WO2014159531A1 - Monoclonal antibodies targeting epcam for detection of prostate cancer lymph node metastases - Google Patents

Monoclonal antibodies targeting epcam for detection of prostate cancer lymph node metastases Download PDF

Info

Publication number
WO2014159531A1
WO2014159531A1 PCT/US2014/024021 US2014024021W WO2014159531A1 WO 2014159531 A1 WO2014159531 A1 WO 2014159531A1 US 2014024021 W US2014024021 W US 2014024021W WO 2014159531 A1 WO2014159531 A1 WO 2014159531A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
seq
cancer
cdr
domain
Prior art date
Application number
PCT/US2014/024021
Other languages
French (fr)
Inventor
Barrett R. Harvey
Kenneth L. PINKSTON
Ali AZHDARINA
Eva M. Sevick-Muraca
Original Assignee
The Board Of Regents Of The University Of Texas System
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Board Of Regents Of The University Of Texas System filed Critical The Board Of Regents Of The University Of Texas System
Priority to US14/774,795 priority Critical patent/US9790274B2/en
Publication of WO2014159531A1 publication Critical patent/WO2014159531A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • A61K47/6865Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from skin, nerves or brain cancer cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1045Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against animal or human tumor cells or tumor cell determinants
    • A61K51/1066Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against animal or human tumor cells or tumor cell determinants the tumor cell being from skin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1093Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody conjugates with carriers being antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3053Skin, nerves, brain
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57434Specifically defined cancers of prostate
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants

Definitions

  • the present invention relates generally to the field of cancer biology. More particularly, it concerns monoclonal antibodies targeting EpCAM antigen.
  • Dual-nuclear and fluorescently labeled or singly fluorescently labeled contrast agents promise the advantage of molecularly-guided surgical resection via surgical field near- infrared fluorescence (NIRF) imaging following (in the case of dual labeled agents) nuclear imaging for general localization.
  • NIRF near- infrared fluorescence
  • LN lymph node
  • PCa prostate-specific antigen
  • a multimodal contrast agent is needed to provide accurate, non- invasive LN staging of PCa and to guide surgical resection of cancer-positive LNs, while sparing resection of cancer-negative LNs and potentially reducing surgical morbidity and improving survivorship.
  • the present invention provides methods and compositions for use in detecting and monitoring epithelial cancer cells.
  • methods and compositions of the embodiments can be used for monitoring the spread of a epithelial cancer from primary tumor sites to the lymphatics and/or for identifying cancer-positive lymph nodes, or for delineating within the surgical field of view, cancer versus cancer free tissues.
  • the present invention is directed towards an isolated or recombinant monoclonal antibody that specifically binds to a EpCAM polypeptide (e.g., a human EpCAM polypeptide).
  • a EpCAM polypeptide e.g., a human EpCAM polypeptide
  • an antibody competes for the binding of an EpCAM polypeptide with the "153" or "7" monoclonal antibody.
  • an antibody of the embodiments can be defined as having an equilibrium 3 ⁇ 4 relative to a human EpCAM polypeptide of less than about 5 nM or 10 nM, e.g., an equilibrium K D of between about 0.1 and 10 nM.
  • the antibody can comprise all or part of the heavy chain variable region and/or the light chain variable region of EpCAM-binding monoclonal antibodies.
  • a polypeptide can comprise an amino acid sequence that corresponds to a first, second, and/or third complementarity determining region (CDR) from the light variable and/or heavy variable chain of a EpCAM monoclonal antibody of the present invention.
  • CDR complementarity determining region
  • an isolated antibody comprises CDR sequences at least 80%, 90% or 95% identical to the CDR regions of the "153" or “7” antibody heavy and light chain amino acid sequences.
  • an antibody comprises CDR regions identical to the 153 or 7, except for one or two amino acid substitutions, deletions or insertions at one or more of the CDRs.
  • the antibody can comprise CDRs wherein the CDR sequences comprise 1 or 2 amino acid substitutions in the VH CDRl, VH CDR2, V H CDR3, V L CDRl, V L CDR2 and/or V L CDR3 relative to the CDRs of a 153 or 7 monoclonal antibody.
  • an antibody of the embodiments comprises (a) a first V H CDR at least 80% identical to V H CDRl of 153 (SEQ ID NO: 1), or 7 (SEQ ID NO: 9); (b) a second V H CDR at least 80% identical to V H CDR2 of 153 (SEQ ID NO: 2), or 7 (SEQ ID NO: 10); (c) a third V H CDR at least 80% identical to V H CDR3 of 153 (SEQ ID NO: 3), or 7 (SEQ ID NO: 1 1); (d) a first V L CDR at least 80% identical to V L CDRl of 153 (SEQ ID NO: 4), or 7 (SEQ ID NO: 12); (e) a second V L CDR at least 80% identical to V L CDR2 of 153 (SEQ ID NO: 5), or 7 (SEQ ID NO: 13); and (f) a third V L CDR at least 80% identical to V L CDR3 of 153 (SEQ ID NO:
  • an antibody of the embodiments comprises each of the CDRs of a 153 antibody, such as, (a) a first V H CDR is identical to SEQ ID NO: 1; (b) a second V H CDR is identical to SEQ ID NO: 2; (c) a third V H CDR is identical to SEQ ID NO: 3; (d) a first V L CDR is identical to SEQ ID NO: 4; (e) a second V L CDR is identical to SEQ ID NO: 5; and (f) a third V L CDR is identical to SEQ ID NO: 6.
  • the isolated antibody comprises a VH domain at least about 80% identical to the VH domain of 153 (SEQ ID NO: 7) and a VL domain at least about 80% identical to the VL domain of 153 (SEQ ID NO: 8).
  • the isolated antibody comprises VH and VL domains identical to or derived from those of monoclonal antibody 153.
  • an antibody of the embodiments comprises each of the CDRs of a 7 antibody, such as, (a) a first VH CDR is identical to SEQ ID NO: 9; (b) a second V H CDR is identical to SEQ ID NO: 10; (c) a third V H CDR is identical to SEQ ID NO: 1 1; (d) a first V L CDR is identical to SEQ ID NO: 12; (e) a second V L CDR is identical to SEQ ID NO: 13; and (f) a third V L CDR is identical to SEQ ID NO: 14.
  • the isolated antibody comprises a VH domain at least about 80% identical to the VH domain of 7 (SEQ ID NO: 15) and a V L domain at least about 80% identical to the V L domain of 7 (SEQ ID NO: 16).
  • the isolated antibody comprises VH and VL domains identical to or derived from those of monoclonal antibody 7.
  • an antibody of the embodiments may be an IgG (e.g., IgGl, IgG2, IgG3 or IgG4), IgM, IgA, or an antigen binding fragment thereof.
  • the antibody may be a Fab', a F(ab')2 a F(ab')3, a monovalent scFv, a bivalent scFv, or a single domain antibody.
  • the antibody may be a human, humanized, or de-immunized antibody.
  • the isolated antibody is the "153" or "7" monoclonal antibody and products derived thereof.
  • an antibody may be conjugated to an imaging agent or a therapeutic agent.
  • the therapeutic agent can be a chemotherapeutic agent, a toxin, or a radionuclide (e.g., a radionuclide sequestered by a chelate).
  • Imagining agents for use according to the embodiments include, without limitation, fluorophores, dyes, a MRI active agent, radionuclides or a fluorescence radionuclide.
  • an antibody can be conjugated to two or more agents, such as to a fluorophore and a radio-imaging agent.
  • a recombinant polypeptide comprising an antibody VH domain comprising CDRs 1-3 of the VH domain of 153 (SEQ ID NOs: 1, 2 and 3); or CDRs 1-3 of the V H domain of 7 (SEQ ID NOs: 9, 10 and 1 1).
  • a recombinant polypeptide is provided comprising an antibody VL domain comprising CDRs 1-3 of the V L domain of 153 (SEQ ID NOs: 4, 5 and 6); or 7 (SEQ ID NOs: 12, 13 and 14).
  • an isolated polynucleotide molecule comprising nucleic acid sequence encoding an antibody or a polypeptide comprising an antibody VH or VL domain disclosed herein.
  • the polynucleotide molecule is an expression vector.
  • a host cell that produces a monoclonal antibody or recombinant polypeptide of the embodiments.
  • the host cell is a mammalian cell, a yeast cell, a bacterial cell, a ciliate cell, or an insect cell.
  • the host cell is a hybridoma cell.
  • a method of manufacturing an antibody of the present embodiments comprising expressing one or more polynucleotide molecule(s) encoding a VL or VH chain of an antibody disclosed herein in a cell and purifying the antibody from the cell.
  • pharmaceutical compositions are provided comprising an antibody or antibody fragment disclosed supra. Such a composition may further comprise a pharmaceutically acceptable carrier and may or may not contain additional active ingredients.
  • a polypeptide of the embodiments comprises an amino acid segment that is at least 80, 85, 90, 95, 96, 97, 98, 99, or 100% identical (or any range derivable therein) to a V, VJ, VDJ, D, DJ, J or CDR domain of a EpCAM-binding antibody (as provided in Table 1).
  • a polypeptide may comprise 1, 2 or 3 amino acid segment that are at least 80, 85, 90, 95, 96, 97, 98, 99, or 100% identical (or any range derivable therein) to CDRs 1, 2, and/or 3 a EpCAM-binding antibody as provided in Table 1.
  • a method of preparing a subject for imaging comprising administering to the subject an effective amount of an antibody of the embodiments, wherein the antibody is conjugated to an imaging agent.
  • a method for imaging a subject comprising (a) administering an effective amount of an antibody of the embodiments to the subject, wherein the antibody is conjugated to an imaging agent; and (b) detecting the imaging agent in the subject.
  • detecting the imaging agent can comprises performing fluorescence imaging, PET, MRI, CT, SPECT or a combination thereof on the subject.
  • images can be acquired using a fluorescence imaging system as described in U.S.
  • a subject for imaging is a subject having as cancer. Accordingly, in aspects, imaging a subject comprises imaging cancer cells in a subject.
  • a method can be defined as a method for detecting cancer metastasis, such as lymphoid metastasis (e.g., lymph node metastasis of a prostate cancer) or for delineating tumor margins and infiltrating cancer within the surgical field of view.
  • cancer metastasis such as lymphoid metastasis (e.g., lymph node metastasis of a prostate cancer) or for delineating tumor margins and infiltrating cancer within the surgical field of view.
  • a method for detecting a cancer in a subject comprising testing for the presence of elevated EpCAM relative to a control in a sample from the subject, wherein the testing comprises contacting the sample with an antibody disclosed herein.
  • the method may be an in vitro or in vivo method.
  • methods that can be used to test a sample for EpCAM expression include, without limitation, ELISA, immunohistochemistry (IHC), fluorescence microscopy, and flow cytometry.
  • a method for treating a subject having a cancer comprising administering an effective amount of an antibody disclosed herein.
  • the antibody is a monoclonal antibody of the present invention, such as 153 or 7 monoclonal antibody, or a recombinant polypeptide comprising antibody segments derived therefrom.
  • the antibody is conjugated to a therapeutic agent, such as a toxin or chemotherapeutic agent.
  • a method may further comprise administering at least a second anticancer therapy to the subject.
  • the second anticancer therapy include, but are not limited to, surgical therapy, chemotherapy, radiation therapy, cryotherapy, hormonal therapy, immunotherapy, or cytokine therapy.
  • Such a second therapy can be administered before, after or essentially simultaneously with an antibody therapeutic of the embodiments.
  • the method may further comprise administering a composition of the present embodiments more than one time to the subject, such as, for example, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20 or more times.
  • a cancer may be a breast cancer, lung cancer, head & neck cancer, prostate cancer, esophageal cancer, tracheal cancer, brain cancer, liver cancer, bladder cancer, stomach cancer, pancreatic cancer, ovarian cancer, uterine cancer, cervical cancer, testicular cancer, colon cancer, rectal cancer, skin cancer, or other epithelial cancer.
  • the cancer is an EpCAM positive cancer, such as a positive prostate cancer.
  • Antibody, therapeutics and imaging agents detailed herein may be administered systemically, locally or distally. In some aspects, an antibody may be administered intravenously, intradermally, intratumorally, intramuscularly, intraperitoneally or subcutaneously.
  • FIG. 1 Dissociation analysis of mAbs from EpCAM/Fc by surface plasmon resonance (SPR) analysis.
  • SPR surface plasmon resonance
  • mAbs and commercial mAb 9601 were captured on a CM5 Sensor chip coupled with goat anti-mouse followed by EpCAM/Fc analyte.
  • Dissociation of EpCAM/Fc from mAb is monitored as a decrease in response units (RU) over time.
  • FIG. 3 Selected candidate mAb binding to native EpCAM expression on PC3 cells compared to that of commercial mAb 9601 via flow cytometry.
  • Anti-EpCAM mAb 7, 153, or 9601 was incubated with PC3 cells followed by detection with PE-labeled secondary antibody.
  • the geometric mean fluorescence intensity (MFI) of the cell population was determined by analysis of 10,000 cells using a BD FACSCalibur Flow Cytometer. The fold increase in MFI for cells incubated with each mAb is shown relative to the MFI of cells incubated with only PE-labeled secondary antibody.
  • MFI Median fluorescence intensity of PC3 cells bound by IRDye- labeled antibody (Ab), as determined by near-infrared (NIR) flow cytometry.
  • NIR near-infrared
  • PC3 cells were analyzed using a customized BD FACS Ariall.
  • FIG. 5 Non-invasive PET/CT and invasive DsRed and near-infrared (NIR) fluorescence images of a representative mouse having a primary prostate tumor and metastatic lymph nodes (LNs). Following dual-labeled mAb administration, coronal (FIG. 5A) and sagittal (FIG. 5B) views from PET/CT show radiotracer signal in the prostate region and several LNs. Regions of interest (ROI, white circles) were drawn around cancer-positive tissue using Inveon Research Workplace (IRW) software. The %ID/g was calculated for each ROI (including background muscle tissue) using IRW. The 3-D view from PET/CT (FIG.
  • FIG. 5C demonstrates better tissue delineation relative to single-slice 2-D views, including delineating primary tumor from bladder, metastatic renal LNs (RLNs) from kidneys and liver (where agent is cleared), and a metastatic sciatic LN (SLN).
  • In situ DsRed fluorescence images (FIGs. 5D and G) of a ventral view from the same animal confirm the presence of a primary prostate tumor and cancer-positive lumbar LNs (LLNs) and RLNs via PC3 cell DsRed reporter-gene expression.
  • the same cancer-positive tissues were detected via in situ NIR fluorescence imaging (FIGs. 5E and H) due to specific binding by dual-labeled mAb (agent).
  • Anatomical location of tissue is depicted by corresponding white light photographs (FIGs. 5F and I) taken during in situ imaging.
  • FIG. 6 Receiver operating characteristic (ROC) curve analyses of TBR of %ID/g measured from PET/CT of LNs in mice given dual-labeled mAb 153, 7, 9601, or 69 (with ⁇ Cu-DOTA and IRDye800). DsRed fluorescence from the gene reporter was considered the true-positive indicator of cancer-positive cells in LNs.
  • the true-positive rate (TPR) or sensitivity was plotted versus false-positive rate (FPR or [1 - specificity]) for each mAb using ascending positive cut-off values, or criteria, that defined positive PET results.
  • the ROC curve and area under the curve (AUC) for each mAb is demonstrated.
  • FIG. 7 Analysis of mAb binding to human and mouse carcinoma cells. Anti-
  • EpCAM mAb 153 or G8.8 was incubated with either human MCF7 or mouse 4T1 cells.
  • the mouse anti-human EpCAM mAb 153 and rat anti-mouse mAb G8.8 were detected with species-specific secondary antibodies, PE-labeled anti-mouse IgG, and DyLIGHT 488- labeled anti-rat IgG, respectively.
  • the geometric mean fluorescence intensity (MFI) was determined with a BD FACSCalibur Flow Cytometer using WinMDI2.9 software.
  • FIGs. 7A and D demonstrate binding specificity of mAb G8.8 to mouse 4T1 cells and no detectable binding to the human cells. Additionally, the entire generated panel of anti-EpCAM mAbs were tested against 4T1 cells and, like mAb 153, demonstrated no detectable binding.
  • FIG. 8 DsRed fluorescence imaging video of animal LN staging. Longitudinal DsRed fluorescence imaging was performed in vivo at two-week intervals post- implantation of DsRed-expressing PC3 cells to monitor primary tumor growth and detect metastatic LNs if present. LN staging could be performed successfully in vivo for some mice, with or without holding the skin slightly taut. Metastases usually involved the lumbar LNs (as shown with holding), and sometimes medial iliac, renal, sciatic, inguinal, or popliteal LNs. [0038] FIG. 9. Comparison of in house generated mAbs 7 and 153 to Veridex mAb
  • VU-1D9 by flow cytometry.
  • FIG. 10 Evaluation of scFv anti-EpCAM binding.
  • MFI mean fluorescent intensities
  • Affinity of anti-EpCAM scFvs was determined by Biacore kinetic analysis.
  • scFv 7 and scFv 153 were measured to have affinities of 4.14 nM and 0.21 nM, respectively (FIGs. 10A and B).
  • FIG. 11 Affinity maturation of lead antibody candidates using APEx directed evolution.
  • FIG. 12 Evaluation of population of 10,000 E. coli cells displaying anti- EpCAM scfv 7 and 153 via the APEx platform can be specifically labeled with EpCAM-Fc followed by detection with fluorescently conjugated secondary antibody with MFIs of 170 and 248, respectively. This is compared to an E. coli population expressing scFv with no affinity to EpCAM as a control, labeled under these conditions with a MFI of only 13. Affinity of scFv correlates with scFv affinity to EpCAM, with resolution dependent on stringency of the wash steps on E. coli prior to evaluation. DESCRIPTION OF ILLUSTRATIVE EMBODIMENTS
  • Epithelial cell adhesion molecule (EpCAM) is overexpressed in many metastasizing epithelial cancers (Spizzo et al, 2006; Spizzo et al, 2004; Went et al, 2006; Fong et al, 2008; Baeuerle et al, 2007) and is an ideal target for dual-labeled agent development.
  • EpCAM in prostatic biopsy samples has been shown through immunohistochemistry (IHC) to be elevated with biochemical recurrence (Benko et al, 201 1) and correlated with Gleason score (Benko et al, 2011 ; Mukherjee et al, 2009), which is an important prognostic factor and is now incorporated into PCa staging nomograms (Edge et al, 2010).
  • IHC immunohistochemistry
  • anti-EpCAM mAb- based imaging agents requires enhanced affinity for improved specificity at sub-therapeutic dosages, elimination of pharmacologic action through ADCC and CDC, and reduced circulation times to improve target-to-background ratios.
  • the inventors developed and quantitatively compared a panel of mAbs specific for human EpCAM, from which high-affinity candidates with specificity for non- overlapping EpCAM epitopes were dual-labeled with a near-infrared (NIR) fluorophore and a conventional chelator/radiotracer, for subsequent assessment via NIRF imaging and quantitative PET/CT for in vivo detection of metastatic LNs in a DsRed-reporter gene mouse model of PCa LN metastasis.
  • NIR near-infrared
  • the inventors sought to first assess whether differences in binding affinity impacted quantitative imaging performance of whole mAb-based contrast agents assessed by receiver operating characteristic (ROC) curve analyses prior to engineering mAb fragment-based imaging agents that will lack properties of therapeutic ADCC and CDC but will have improved imaging pharmacokinetics.
  • ROC receiver operating characteristic
  • MAbs 7 and 153 performed with greater sensitivity during ⁇ /CT when compared to commercial mAb and yielded strong correlations between qualitative NIRF and DsRed-reporter gene imaging.
  • Qualitative comparison of NIRF imaging demonstrated similar results to those found from DsRed reporter-gene imaging and ⁇ /CT.
  • MAbs 7 and 153 are attractive candidates for further agent optimization aimed at enhancing sensitivity and specificity for detection of metastatic LNs in PCa and creating Ab fragments to reduce circulation times and eliminate Fc binding regions responsible for therapeutic action and nonspecific binding.
  • EpCAM has been a putative cancer therapeutic target for decades, this is one of the first efforts to specifically engineer the detection agent for diagnostic use. This could be used as an intraoperative means for surgeons to identify and specifically remove cancer positive lymph nodes during prostatectomy surgery and subsequent lymphadectomy. Additionally, for CTC detection, with the Veridex FDA approved platform, a clear association has been made with CTC quantification and lifespan of patient, demonstrating value of this technology in determining direction of patient treatment (Wallwiener et ah, 2013; Young et ah, 2012; Sun et ah, 2012). The next logical step is to improve the anti- EpCAM agent affinity as it is the primary means of CTC isolation. Improvements in affinity should translate into improved CTC detection while minimizing the blood sample volume necessary from the patient.
  • the terms “treat,” “treating,” “treatment” and “therapy” contemplate an action that occurs while a patient is suffering from epithelial cancers and related disorders that reduces the severity of one or more symptoms or effects of epithelial cancers, metastisis or a related disease or disorder, such as but not limited to breast cancer, lung cancer, head & neck cancer, prostate cancer, esophageal cancer, tracheal cancer, brain cancer, liver cancer, bladder cancer, stomach cancer, pancreatic cancer, ovarian cancer, uterine cancer, cervical cancer, testicular cancer, colon cancer, rectal cancer, skin cancer, or other epithelial cancer and metastasis associated with such.
  • the cancer is an EpCAM positive cancer, such as a positive prostate cancer.
  • the terms “treat,” “treating,” and “treatment” also refers to actions taken toward ensuring that individuals at increased risk of epithelial cancer - metastasis are able to receive appropriate surgical and/or other medical intervention prior to onset of epithelial cancers and disorders.
  • the terms “prevent,” “preventing,” and “prevention” contemplate an action that occurs before a patient begins to suffer from epithelial cancers and related disorders that delays the onset of, and/or inhibits or reduces the severity of, epithelial cancers and related disorders.
  • the terms “manage,” “managing,” and “management” encompass preventing, delaying, or reducing the severity of a recurrence of epithelial cancers, metastisis and related disorders in a patient who has already suffered from such a disease or condition.
  • the terms encompass modulating the threshold, development, and/or duration of the epithelial cancers, metastisis and related disorders or changing how a patient responds to the epithelial cancers, metastisis and related disorders.
  • a "therapeutically effective amount" of a compound is an amount sufficient to provide any therapeutic benefit in the treatment or management of epithelial cancers, metastisis and related disorders or to delay or minimize one or more symptoms associated with epithelial cancers, metastisis and related disorders.
  • a therapeutically effective amount of a compound means an amount of the compound, alone or in combination with one or more other therapies and/or therapeutic agents, which provides any therapeutic benefit in the treatment or management of epithelial cancers, metastisis and related disorders, or related diseases or disorders.
  • therapeutically effective amount can encompass an amount that alleviates epithelial cancers, metastisis and related disorders, improves or reduces epithelial cancers, metastisis and related disorders, improves overall therapy, or enhances the therapeutic efficacy of another therapeutic agent.
  • the “therapeutically effective amount” can be identified at an earlier stage with parameters of 1 epithelial cancers, metastisis and related epithelial cancers, metastisis and related function as identified herein.
  • lymphatic structure(s) refers to all or a portion of structures that make up a mammalian lymphatic system including without limitation, lymph nodes, collecting vessels, lymph trunks, lymph ducts, capillaries, or combinations thereof.
  • the architecture of the lymphatic structures can be described by tortuosity, density, dilation, and other parameters.
  • near-infrared refers to electromagnetic radiation at wavelengths ranging from about 750 nm to about 900 nm.
  • the term "functional imaging" of lymph structures refers to how the structures function in terms of update of dye, the lymphatic flow as determined by the dye, dynamics of flow, and direction of flow of lymph and the associated materials carried by it.
  • the function of the lymphatic structures can be described by lymph velocity, period or frequency of propulsive events, permeability, and other parameters that provide evidence of dysfunction in comparison to normal function imaged in healthy control animals or human subjects. If lymph vessels are "functional" they transport materials, and imaging methods disclosed herein describe this.
  • EpCAM a 40 kD type I transmembrane glycoprotein involved in cell-to-cell interactions and adhesion, is stably overexpressed by most types of epithelial cancers. EpCAM was the first identified tumor-associated antigen that today is known to be intensely and uniformly expressed on epithelial carcinomas while being less intensely expressed, sequestered, and less accessible on the basolateral cell surface of normal epithelia, making it a useful marker for diagnostic applications.
  • EpCAM epithelial cell adhesion molecule
  • an antibody or a fragment thereof that binds to at least a portion of EpCAM protein and its associated use in diagnosis and imaging of diseases are contemplated.
  • the term "antibody” is intended to refer broadly to any immunologic binding agent, such as IgG, IgM, IgA, IgD, and IgE as well as polypeptides comprising antibody CDR domains that retain antigen binding activity.
  • the antibody may be selected from the group consisting of a chimeric antibody, an affinity matured antibody, a polyclonal antibody, a monoclonal antibody, a humanized antibody, a human antibody, or an antigen-binding antibody fragment or a natural or synthetic ligand.
  • the anti- EpCAM antibody is a monoclonal antibody or a humanized antibody.
  • polyclonal or monoclonal antibodies, antibody fragments, and binding domains and CDRs may be created that are specific to EpCAM protein, one or more of its respective epitopes, or conjugates of any of the foregoing, whether such antigens or epitopes are isolated from natural sources or are synthetic derivatives or variants of the natural compounds.
  • antibody fragments suitable for the present invention include, without limitation: (i) the Fab fragment, consisting of VL, VH, CL, and CHI domains; (ii) the "Fd” fragment consisting of the VH and CHI domains; (iii) the "Fv” fragment consisting of the VL and VH domains of a single antibody; (iv) the "dAb” fragment, which consists of a VH domain; (v) isolated CDR regions; (vi) F(ab')2 fragments, a bivalent fragment comprising two linked Fab fragments; (vii) single chain Fv molecules ("scFv”), wherein a VH domain and a VL domain are linked by a peptide linker that allows the two domains to associate to form a binding domain; (viii) bi-specific single chain Fv dimers (see U.S.
  • Fv, scFv, or diabody molecules may be stabilized by the incorporation of disulphide bridges linking the VH and VL domains.
  • Minibodies comprising a scFv joined to a CH3 domain may also be made (Hu et ah, 1996).
  • Antibody-like binding peptidomimetics are also contemplated in embodiments. Liu et al. (2003) describe "antibody like binding peptidomimetics" (ABiPs), which are peptides that act as pared-down antibodies and have certain advantages of longer serum half-life as well as less cumbersome synthesis methods.
  • Animals may be inoculated with an antigen, such as purified peptide or cells expressing the antigen on their surface, in order to produce antibodies specific for EpCAM protein. Frequently an antigen is bound or conjugated to another molecule to enhance the immune response.
  • a conjugate is any peptide, polypeptide, protein, or non- proteinaceous substance bound to an antigen that is used to elicit an immune response in an animal.
  • Antibodies produced in an animal in response to antigen inoculation comprise a variety of non-identical molecules (polyclonal antibodies) made from a variety of individual antibody producing B lymphocytes.
  • a polyclonal antibody is a mixed population of antibody species, each of which may recognize a different epitope on the same antigen. Given the correct conditions for polyclonal antibody production in an animal, most of the antibodies in the animal's serum will recognize the collective epitopes on the antigenic compound to which the animal has been immunized. This specificity is further enhanced by affinity purification to select only those antibodies that recognize the antigen or epitope of interest.
  • a monoclonal antibody is a single species of antibody wherein every antibody molecule recognizes the same epitope because all antibody producing cells are derived from a single B-lymphocyte cell line.
  • the methods for generating monoclonal antibodies generally begin along the same lines as those for preparing polyclonal antibodies.
  • rodents such as mice and rats are used in generating monoclonal antibodies.
  • rabbit, sheep, or frog cells are used in generating monoclonal antibodies. The use of rats is well known and may provide certain advantages.
  • Mice e.g., BALB/c mice) are routinely used and generally give a high percentage of stable fusions.
  • Hybridoma technology involves the fusion of a single B lymphocyte from a mouse previously immunized with a EpCAM antigen with an immortal myeloma cell (usually mouse myeloma).
  • This technology provides a method to propagate a single antibody-producing cell for an indefinite number of generations, such that unlimited quantities of structurally identical antibodies having the same antigen or epitope specificity (monoclonal antibodies) may be produced.
  • the antibody is a chimeric antibody, for example, an antibody comprising antigen binding sequences from a non-human donor grafted to a heterologous non-human, human, or humanized sequence (e.g., framework and/or constant domain sequences).
  • a heterologous non-human, human, or humanized sequence e.g., framework and/or constant domain sequences.
  • Methods have been developed to replace light and heavy chain constant domains of the monoclonal antibody with analogous domains of human origin, leaving the variable regions of the foreign antibody intact.
  • "fully human" monoclonal antibodies are produced in mice transgenic for human immunoglobulin genes.
  • Methods have also been developed to convert variable domains of monoclonal antibodies to more human form by recombinantly constructing antibody variable domains having both rodent, for example, mouse, and human amino acid sequences.
  • Antibodies may be produced from any animal source, including birds and mammals.
  • the antibodies are ovine, murine (e.g., mouse and rat), rabbit, goat, guinea pig, camel, horse, or chicken.
  • newer technology permits the development of and screening for human antibodies from human combinatorial antibody libraries.
  • bacteriophage antibody expression technology allows specific antibodies to be produced in the absence of animal immunization, as described in U.S. Pat. No. 6,946,546, which is incorporated herein by reference. These techniques are further described in: Marks (1992); Stemmer (1994); Gram et al. (1992); Barbas et al. (1994); and Schier et al. (1996).
  • antibodies to EpCAM will have the ability to bind EpCAM regardless of the animal species, monoclonal cell line, or other source of the antibody.
  • Certain animal species may be less preferable for generating diagnostic imaging antibodies because they may be more likely to cause allergic response due to activation of the complement system through the "Fc" portion of the antibody.
  • whole antibodies may be enzymatically digested into "Fc" (complement binding) fragment, and into antibody fragments having the binding domain or CDR. Removal of the Fc portion reduces the likelihood that the antigen antibody fragment will elicit an undesirable immunological response, and thus, antibodies without Fc may be preferential for prophylactic or therapeutic treatments.
  • antibodies may also be constructed so as to be chimeric or partially or fully human, so as to reduce or eliminate the adverse immunological consequences resulting from administering to an animal an antibody that has been produced in, or has sequences from, other species.
  • Substitutional variants typically contain the exchange of one amino acid for another at one or more sites within the protein, and may be designed to modulate one or more properties of the polypeptide, with or without the loss of other functions or properties. Substitutions may be conservative, that is, one amino acid is replaced with one of similar shape and charge.
  • Conservative substitutions are well known in the art and include, for example, the changes of: alanine to serine; arginine to lysine; asparagine to glutamine or histidine; aspartate to glutamate; cysteine to serine; glutamine to asparagine; glutamate to aspartate; glycine to proline; histidine to asparagine or glutamine; isoleucine to leucine or valine; leucine to valine or isoleucine; lysine to arginine; methionine to leucine or isoleucine; phenylalanine to tyrosine, leucine or methionine; serine to threonine; threonine to serine; tryptophan to tyrosine; tyrosine to tryptophan or phenylalanine; and valine to isoleucine or leucine.
  • substitutions may be non-conservative such that a function or activity of the polypeptide is affected.
  • Non-conservative changes typically involve substituting a residue with one that is chemically dissimilar, such as a polar or charged amino acid for a nonpolar or uncharged amino acid, and vice versa.
  • Proteins may be recombinant, or synthesized in vitro.
  • a non- recombinant or recombinant protein may be isolated from bacteria. It is also contemplated that a bacteria containing such a variant may be implemented in compositions and methods. Consequently, a protein need not be isolated.
  • An antibody or preferably an immunological portion of an antibody can be chemically conjugated to, or expressed as, a fusion protein with other proteins.
  • a fusion protein with other proteins.
  • all such fused proteins are included in the definition of antibodies or an immunological portion of an antibody.
  • Embodiments provide antibodies and antibody-like molecules against EpCAM, polypeptides and peptides that are linked to at least one agent to form an antibody conjugate or payload.
  • it is conventional to link or covalently bind or complex at least one desired molecule or moiety.
  • a molecule or moiety may be, but is not limited to, at least one effector or reporter molecule.
  • Effector molecules comprise molecules having a desired activity, e.g., cytotoxic activity.
  • Non-limiting examples of effector molecules that have been attached to antibodies include toxins, therapeutic enzymes, antibiotics, radio-labeled nucleotides and the like.
  • reporter molecule is defined as any moiety that may be detected using an assay.
  • reporter molecules that have been conjugated to antibodies include enzymes, radiolabels, haptens, fluorescent labels, phosphorescent molecules, chemiluminescent molecules, chromophores, luminescent molecules, photoaffinity molecules, colored particles or ligands, such as biotin.
  • Some attachment methods involve the use of a metal chelate complex employing, for example, an organic chelating agent such a diethylenetriaminepentaacetic acid anhydride (DTPA); ethylenetriaminetetraacetic acid; N- chloro-p-toluenesulfonamide; and/or tetrachloro-3-6?-diphenylglycouril-3 attached to the antibody.
  • organic chelating agent such as diethylenetriaminepentaacetic acid anhydride (DTPA); ethylenetriaminetetraacetic acid; N- chloro-p-toluenesulfonamide; and/or tetrachloro-3-6?-diphenylglycouril-3 attached to the antibody.
  • Monoclonal antibodies may also be reacted with an enzyme in the presence of a coupling agent such as glutaraldehyde or periodate. Conjugates with fluorescein markers are prepared in the presence of these coupling agents or by reaction with an isothi
  • the conjugation of the radionuclide to the antibodies or fragments of the invention can be made using any conventional techniques such as the use of a linker between the antibody and the radioisotope.
  • the radioimmunoconjugate has a specific activity of from about 0.5 to about 15 mCi/mg, depending on the radionuclide, and may be administered via an intravenous or other route.
  • an antibody or antibody fragment can be detectably labeled through the use of radioisotopes, affinity labels (e.g., biotin, avidin, etc.), fluorescent labels, paramagnetic atoms, etc.
  • affinity labels e.g., biotin, avidin, etc.
  • fluorescent labels e.g., fluorescent labels
  • paramagnetic atoms etc.
  • the detection of foci of such detectably labeled antibodies or antibody fragments might be indicative of a site of tumor development.
  • this technique is done in a non-invasive manner through the use of magnetic imaging, fluorography, PET, etc.
  • Such a diagnostic test may be employed in monitoring the success of treatment of diseases, where presence or absence of EpCAM-positive cells is a relevant indicator.
  • Such imaging may also be employed during surgical resection of lymph nodes comprising metastatic disease to guide said surgical resection.
  • compositions may comprise, for example, at least about 0.1% of an active compound.
  • an active compound may comprise between about 2% to about 75% of the weight of the unit, or between about 25% to about 60%, for example, and any range derivable therein.
  • the pharmaceutical compositions of the present invention are advantageously administered in the form of injectable compositions either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid prior to injection may also be prepared. These preparations also may be emulsified.
  • phrases "pharmaceutical or pharmacologically acceptable” refers to molecular entities and compositions that do not produce an adverse, allergic, or other untoward reaction when administered to an animal, such as a human, as appropriate.
  • the preparation of a pharmaceutical composition comprising an antibody or additional active ingredient will be known to those of skill in the art in light of the present disclosure.
  • animal (e.g., human) administration it will be understood that preparations should meet sterility, pyrogenicity, general safety, and purity standards as required by FDA Office of Biological Standards.
  • aqueous solvents e.g., water, alcoholic/aqueous solutions, saline solutions, parenteral vehicles, such as sodium chloride, Ringer's dextrose, etc.
  • non-aqueous solvents e.g., propylene glycol, polyethylene glycol, vegetable oil, and injectable organic esters, such as ethyloleate
  • dispersion media coatings, surfactants, antioxidants, preservatives (e.g., antibacterial or antifungal agents, anti-oxidants, chelating agents, and inert gases), isotonic agents, absorption delaying agents, salts, drugs, drug stabilizers, gels, binders, excipients, disintegration agents, lubricants, sweetening agents, flavoring agents, dyes, fluid and nutrient replenishers, such like materials and combinations thereof, as would be known to one of ordinary skill in the art.
  • aqueous solvents e.g., water, alcoholic/aqueous solutions,
  • a pharmaceutically acceptable carrier is particularly formulated for administration to a human, although in certain embodiments it may be desirable to use a pharmaceutically acceptable carrier that is formulated for administration to a non-human animal but that would not be acceptable (e.g., due to governmental regulations) for administration to a human. Except insofar as any conventional carrier is incompatible with the active ingredient, its use in the therapeutic or pharmaceutical compositions is contemplated.
  • unit dose refers to physically discrete units suitable for use in a subject, each unit containing a predetermined quantity of the diagnositc composition calculated to produce the desired responses discussed above in association with its administration, i.e., the appropriate route and treatment regimen.
  • the quantity to be administered depends on the effect desired.
  • the actual dosage amount of a composition of the present invention administered to a patient or subject can be determined by physical and physiological factors, such as body weight, the age, health, and sex of the subject, the type of disease being imaged, the extent of disease penetration, previous or concurrent therapeutic interventions, idiopathy of the patient, the route of administration, and the potency, stability, and toxicity of the particular therapeutic substance.
  • a dose may also comprise from about 1 ⁇ g/kg/body weight to about 1000 mg/kg/body weight (this such range includes intervening doses) or more per administration, and any range derivable therein.
  • a derivable range from the numbers listed herein, a range of about 5 ⁇ g/kg/body weight to about 100 mg/kg/body weight, about 5 ⁇ g/kg/body weight to about 500 mg/kg/body weight, etc., can be administered.
  • the practitioner responsible for administration will, in any event, determine the concentration of active ingredient(s) in a composition and appropriate dose(s) for the individual subject.
  • the active compounds can be formulated for parenteral administration, e.g., formulated for injection via the intravenous, intramuscular, sub-cutaneous, or even intraperitoneal routes.
  • parenteral administration e.g., formulated for injection via the intravenous, intramuscular, sub-cutaneous, or even intraperitoneal routes.
  • such compositions can be prepared as either liquid solutions or suspensions; solid forms suitable for use to prepare solutions or suspensions upon the addition of a liquid prior to injection can also be prepared; and, the preparations can also be emulsified.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil, or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form must be sterile and must be fluid to the extent that it may be easily injected. It also should be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • the proteinaceous compositions may be formulated into a neutral or salt form.
  • Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like.
  • a pharmaceutical composition can include a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils.
  • a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils.
  • the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion, and by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • kits are envisioned containing diagnostic agents and/or other therapeutic and delivery agents.
  • the present invention contemplates a kit for preparing and/or administering a diagnostic agent of the invention.
  • the kit may comprise one or more sealed vials containing any of the pharmaceutical compositions of the present invention.
  • the kit may include, for example, at least one EpCAM antibody as well as reagents to prepare, formulate, and/or administer the components of the invention or perform one or more steps of the inventive methods.
  • the kit may also comprise a suitable container, which is a container that will not react with components of the kit, such as an eppendorf tube, an assay plate, a syringe, a bottle, or a tube.
  • the container may be made from sterilizable materials such as plastic or glass.
  • the kit may further include an instruction sheet that outlines the procedural steps of the methods set forth herein, and will follow substantially the same procedures as described herein or are known to those of ordinary skill in the art.
  • the instruction information may be in a computer readable media containing machine-readable instructions that, when executed using a computer, cause the display of a real or virtual procedure of delivering a pharmaceutically effective amount of a diagnostic agent.
  • mice were immunized with either recombinant human (rhu) EpCAM/Fc (960-EP, R&D Systems, Minneapolis, MN) or MCF7 cells (HTB-22TM, ATCC, Manassas, VA), a human breast adenocarcinoma line that expresses EpCAM (Prang et ah, 2005; Munz et ah, 2010).
  • EpCAM /Fc was administered using standard methods as previously described (Pinkston et ah, 2011).
  • MCF7 cells were grown in GIBCO® Dulbecco's Minimal Essential Medium (DMEM) (Invitrogen Corporation, Carlsbad, CA) with 10% fetal bovine serum (FBS) to 80%-100% confluency in T75 flasks, detached with Trypsin- ethylenediaminetetraacetic acid (EDTA) (Cellgro, Manassas, VA), washed in phosphate buffered saline (PBS), pH 7.4, and injected intraperitoneally (ip) at 10 7 cells per mouse every ten days for five injections.
  • DMEM Dulbecco's Minimal Essential Medium
  • FBS fetal bovine serum
  • EDTA Trypsin- ethylenediaminetetraacetic acid
  • PBS phosphate buffered saline
  • ip intraperitoneally
  • Mouse splenocytes were collected and fused with SP2/0 mouse myeloma cells (Pinkston et ah, 2011).
  • MAbs from parental wells were evaluated for binding specificity to rhuEpCAM/Fc via an enzyme-linked immunosorbent assay (ELISA) followed by kinetic screening to rank order high affinity clones using surface plasmon resonance (SPR) (Canziani et al, 2004).
  • ELISA enzyme-linked immunosorbent assay
  • SPR surface plasmon resonance
  • each selected mAb from the inventor's panel was added to the plate at 2 ⁇ g/ml and incubated for 30 min at room temperature (RT). This was followed by the addition of HRP -conjugated mAb at 0.2 ⁇ g/ml and incubation for 30 min at RT.
  • HRP-conjugated mAb was detected via reaction with 3, 3', 5,5'- tetramethylbenzidine (TMB) substrate (Sigma-Aldrich, St.
  • the pass-through material containing fragment antigen-binding (Fab) regions was dialyzed against PBS and retained for enrichment via size exclusion chromatography.
  • a GE ⁇ Explorer GE Healthcare, Piscataway, NJ
  • fast protein liquid chromatography FPLC
  • HR 10/30 FPLC column GE Healthcare
  • Purity was then assessed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE).
  • HBS-EP+ (10 mM HEPES, 150 mM NaCl, 3 mM EDTA, 0.05% v/v Tween 20, pH 7.4) as running buffer.
  • EpCAM/Fc ligand (R&D Systems) was diluted in HBS-EP+ to 0.5 ⁇ g/ml and applied to the coated chip surface. Fab fragments were then tested at five different molar concentrations starting at 50 nM after performing a two-fold dilution series. Flow cells were regenerated with 100 mM phosphoric acid. All samples were applied in duplicate.
  • PC3 cells human prostate adenocarcinoma line, CRL-1435TM, ATCC, Manassas, VA
  • PC3 cells grown to 80%- 100% confluency in DMEM with 10% FBS
  • Trypsin-EDTA and resuspended in PBS containing 2% bovine serum albumin (BSA).
  • BSA bovine serum albumin
  • phycoerythrin (PE)-conjugated donkey F(ab')2 anti-mouse IgG (Jackson ImmunoResearch, West Grove, PA) at 5 ⁇ g per sample was added for an additional 20-min incubation.
  • Cells were washed again with PBS-BSA and fixed in 2% paraformaldehyde prior to analyzing (10,000 events) on a BD FACSCalibur Flow Cytometer (BD Biosciences, San Jose, CA). Analyses of mean fluorescence intensities (MFI) were performed using WinMDI2.9 software.
  • IgG 2b isotype control mAb (BD Pharmingen, San Diego, CA) and an in-house generated IgGi isotype control mAb were used in each assay to test for nonspecific binding. After centrifugation at 350 x g for 5 min at RT, supernatants were discarded and cells were washed with PBS. As a positive control, IRDye-labeled secondary Ab, goat anti-mouse IgG (LI-COR), was then added to reactions containing cells incubated with unlabeled, commercial mAb 9601. As a negative control for secondary Ab staining, the secondary Ab was also added to cells without primary Ab.
  • IRDye-labeled secondary Ab goat anti-mouse IgG (LI-COR)
  • Dual-labeled mAbs (40-50 ⁇ g/7.4-12.95 MBq) were administered via the tail vein at 10-12 weeks post-implantation. Within 18-24 hr, non-invasive ⁇ /CT was performed and immediately followed by in vivo and in situ DsRed reporter-gene and NIRF optical imaging.
  • PET, NIRF, and DsRed Fluorescence Imaging Fluorescence imaging was conducted using custom-built instrumentation described elsewhere (Hall et ah, 201 1).
  • excitation light was supplied at 568 nm by a tunable, air- cooled, argon/krypton laser and emission light (610 ⁇ 5 nm) was collected through a bandpass filter (Andover Corp., Salem, NH).
  • NIRF imaging utilized excitation light at 786 nm from a laser diode and emission light was collected through two tandem bandpass filters collecting light at 830 ⁇ 5 nm (Zhu et ah, 2010).
  • Non-invasive ⁇ /CT was performed using an Inveon System (Siemens Medical Solutions, Knoxville, TN) with multimodality CT and docked PET. Images were analyzed and %ID/g was quantified from PET images using Inveon Research Workplace software (Siemens Medical Solutions). Target-to-background ratios (TBRs) were computed from the %ID/g normalized to normal muscle background. Previously, the inventors showed that LN DsRed fluorescence measured in situ or ex vivo provided comparable or better true- positive LN discrimination than pathology (Hall et al, 2010).
  • the inventors used in situ or ex vivo DsRed fluorescence as a measure of ground truth for LN cancer positivity and computed the PET true-positive rate (TPR or sensitivity) versus false-positive rate (FPR, or [1 - specificity]) of all LNs examined using ascending TBR as positive cut-off criteria during ROC curve analyses.
  • the area under the curve (AUC) was then computed for each dual-labeled mAb tested.
  • Anti-EpCAM MAbs Selected by Off Rate Analysis and Competitive Binding. Screening and analyses of single-cell clones from parental wells of hybridoma fusions were performed as previously described (Pinkston et al, 20 ⁇ ⁇ ; Canziani et al, 2004) and yielded a mAb panel with slower off rates than that of commercial mAb 9601 (FIG. 1). During competitive binding assays, binding of some mAbs, including mAbs 17, 73, 81, 144, and 153, to EpCAM blocked binding by mAb 9601, suggesting shared binding epitopes (FIG. 2A).
  • mAbs 7, 18, 33, 38, 66, 70, 83, 85, 124, 133, and 138 did not inhibit mAb 9601 binding, suggesting epitopes that do not overlap or binding sites that do not compete with that of mAb 9601 (FIG. 2B).
  • the inventors found that mAbs that do not have overlapping epitope binding with mAb 9601, have an overlapping footprint with mAb 7.
  • the mAb panel could be categorized into two unique groups with specificity for two separate, non-overlapping epitopes on the EpCAM protein.
  • MAbs from each group, mAbs 153 and 7, were chosen for further evaluation.
  • the VH and VL sequences, including the corresponding CDR sequences, for mAb 153 and mAb 7 are shown in Table 1. Table 1. VH and VL CDR Sequences for mAbs 153 and 7.
  • Fab fragments of mAb 153 demonstrated a slower Ka or "off rate than that of mAb 7, which in turn was slower than that of mAb 9601 Fab fragment, a trend consistent with SPR analysis of dissociation avidity of captured IgG antibody using the bivalent EpCAM/Fc as the analyte (Table 2).
  • both mAb 153 and 7 Fab fragments demonstrated significantly faster "on" rates (K a ) than that of mAb 9601.
  • the equilibrium affinity constant (3 ⁇ 4) was found to be 15.4, 0.54, and 2.3 nM for mAbs 9601, 153, and 7 Fab fragments, respectively. These affinity constants translated to a 28- and 7-fold increase in affinity of mAbs 153 and 7 above mAb 9601, respectively.
  • IgGi isotype control mAb 69 had a significantly lower MFI (124.7 ⁇ 7.3 MFI units) relative to mAbs 7 and 153 (P ⁇ 0.05), as did commercial IgG 2b isotype control mAb (149 ⁇ 6.0 MFI units) relative to mAb 9601.
  • the biological activity, measured as the mean percentage of PC3 cells stained, of conjugated mAb 7 (86.4 ⁇ 1.8%) was significantly greater than that of IRDye-labeled mAb 9601 (79.5 ⁇ 0.8%) (P ⁇ 0.05, Table 2). While the biological activity of mAb 153 (81.8 ⁇ 1.5%) was also higher than that of mAb 9601, the difference was not found to be significant.
  • the percentage of cells stained by IgGi isotype control mAb 69 was significantly low relative to mAbs 7 and 153, as was the percentage of cells stained by commercial IgG 2b isotype control mAb (0.7 ⁇ 0.1%) relative to mAb 9601 (P ⁇ 0.05).
  • the MFI (1 1 16.5 ⁇ 54.5 MFI units) as well as the percentage (80.2 ⁇ 5.5%) of cells bound by unlabeled mAb 9601 followed by incubation with IRDye-labeled secondary Ab was not significantly different from that of cells stained directly by conjugated mAb 9601, demonstrating that the NIR fluorophore label did not significantly hinder mAb binding to its target on PC3 cells.
  • ⁇ NA is an isotype control targeting pili.
  • Non-invasive, DsRed fluorescence imaging of each mouse at two-week intervals allowed for monitoring of primary prostate tumors and detection of LN involvement in some animals. While primary tumors and some metastatic LNs could be easily visualized, it was necessary to hold the abdominal skin taut on some mice for LN staging (see FIG. 8). By week 10-12 post-implantation, metastases usually involved the lumbar LNs, and sometimes also medial iliac, renal, sciatic, inguinal, or popliteal LNs.
  • radiotracer signal from primary prostatic tumors and LNs bound by dual-labeled mAb was detected.
  • FIG. 5 shows coronal (FIG.
  • FIG. 5A sagittal (FIG. 5B), and 3-D (FIG. 5C) views from ⁇ /CT of a representative mouse in which the prostate, lumbar, and renal LNs, and one sciatic LN were PET-positive.
  • In situ DsRed gene-reporter imaging demonstrated the presence of PC3 cells in the same tissues, confirming a primary tumor and LN metastases. DsRed fluorescence in this animal's right sciatic LN was observed during in vivo imaging of the dorsal side as well as ex vivo imaging.
  • in situ NIRF imaging of the same mouse FIGs.
  • FIG. 6 shows the ROC curve generated for each dual-labeled mAb tested.
  • the AUC for dual-labeled mAbs 7 (86.8%) and 153 (78.0%) were both significantly greater than the 50% nondiscriminatory line (P ⁇ 0.05), whereas the AUC for mAb 9601 (60.7%) was not significantly different from a 50/50 random guess (Table 3).
  • the conversion of the lead mAbs to scFvs with comparable affinities to the parent antibody binding domain (i) demonstrates the capability of accurately measuring and retaining affinity while converting to different antibody formats and (ii) the conversion to the scFv antibody format allows for the expression of the antibody binding domain in Escherichia coli, required for affinity maturation of this antibody using display technologies.
  • Secreted mAbs will then be purified from medium using protein A/G affinity chromatography. The concentration of purified mAbs will be determined by OD280 nm and the purity by SDS-PAGE. Biacore analysis will confirm affinity improvement of IgG as described (Hall et al, 2012).
  • scFv 7 and scFv 153 DNA sequences that have been identified will be utilized as template to generate mutagenic libraries as previously described (Fromant et al. 1995). Each sequence resulting from mutagenesis that bears a unique set of mutations is then cloned into the pAPExl vector for protein expression and display of the library on the E. coli inner membrane via a new lipoprotein A (NlpA) fusion as previously described (Harvey et al, 2004; Harvey et al, 2006).
  • NlpA new lipoprotein A
  • Sentinel LN dissection has been standard-of-care in breast cancer staging and recently, has been under investigation for staging PCa (Holl et al, 2009; Janetschek et al, 2007). It is also notable that with the advent of improved systemic treatment, recent studies have shown that breast cancer patients with limited sentinel LN metastases have gained no benefit from extensive LN dissection (Giuliano et al, 2011). Earlier detection of PCa using PSA testing and PLND may continue to decrease mortality rates due to PCa, but may also inherently increase the risk for overtreatment in low-risk PCa patients (Jemal et al, 2010), especially with the advent of new pharmacologic therapies against PCa.
  • a molecularly targeted imaging agent that offers both benefits of noninvasive imaging and intraoperative guidance for resection of cancer-positive LNs is needed for more accurate and effective PCa staging as well as staging of other epithelial cancers.
  • EpCAM which is overexpressed in >98% of PCa cases (Went et al,
  • anti-EpCAM mAbs with attenuated affinities exhibit less systemic toxicity and have been moderately successful in clinical trials (Munz et ah, 2010). Yet, when administered as a diagnostic at sub-therapeutic dosages, the attenuated affinity of anti-EpCAM mAbs could potentially impact imaging sensitivity, thus motivating the inventor's studies to evaluate imaging performance. Furthermore, when full-length, affinity-attenuated mAbs are converted to fragments, their reduced circulation times will not only offer more desirable pharmacokinetics to reduce background imaging signal, but would most likely result in reduced targeting and imaging sensitivity. While others have employed therapeutic mAbs as radio- and fluorescently- labeled diagnostic agents in clinical and preclinical studies, the inventors sought herein to first develop mAbs with the highest affinities and assess imaging performance prior to further modification.
  • the inventor's SPR-based selection strategy allowed us to identify high-affinity mAbs produced from hybridoma clones for testing in a mouse model of human PCa LN metastases. Imaging analysis was accelerated by the use of a fluorescent DsRed gene reporter as "ground truth" during assessment of LN metastases. Consistent with SPR affinity of the Fab fragment measurements, dual-labeled mAbs 153 and 7 as imaging agents possessed greater sensitivity than mAb 9601 and had a significantly higher AUC than the 50% line of non-discrimination. In contrast, the AUC of mAb 9601 was not statistically greater than the line of random chance.
  • Reduction of nonspecific binding can be achieved through modification of the Fc region (Strohl et al , 2009) or by developing dual-labeled specific Ab fragments, such as diabodies, or single-chain variable fragments (scFv) to eliminate Fc regions (Olafsen et al, 2010).
  • Fc receptor-mediated interaction with mAb is known to play an important role in reducing mAb clearance (Kosterink et al, 2007), increasing half-life in the blood circulation (Datta-Mannan et al, 2007), and therefore increasing background signal that in turn can reduce imaging contrast.
  • these phenomena obviate the need for mAb fragment-based agent development during contrast agent optimization for detection of metastatic LNs in PCa.
  • Pilepich et al Surgical staging in carcinoma of the prostate: the RTOG experience. Radiation Therapy Oncology Group. Prostate. 1984;5(5):471-476. Musch et al, Complications of pelvic lymphadenectomy in 1,380 patients undergoing radical retropubic prostatectomy between 1993 and 2006. J Urol. Mar 2008; 179(3):923-928; discussion 928-929.
  • Ep-CAM epithelial cell adhesion molecule
  • EpCAM epithelial cell adhesion molecule

Abstract

Embodiments of the invention provide high affinity recombinant EpCAM-binding antibodies. Methods of using a such antibodies as imaging agents, diagnostics and therapeutics are also provided. Dual-nuclear and fluorescently labeled or singly fluorescently labeled contrast agents promise the advantage of molecularly-guided surgical resection via surgical field near-infrared fluorescence (NIRF) imaging following (in the case of dual labeled agents) nuclear imaging for general localization. Currently, nodal staging of most cancers is performed following lymph node (LN) biopsy and dissection for subsequent pathological examination.

Description

DESCRIPTION
MONOCLONAL ANTIBODIES TARGETING EPCAM FOR DETECTION OF
PROSTATE CANCER LYMPH NODE METASTASES
[0001] This application claims the benefit of United States Provisional Patent Application No 61/782,601, filed March 14, 2013, the entirety of which is incorporated herein by reference.
[0002] The invention was made with government support under Grant No. U54 CA 136404 awarded by the National Institutes of Health. The government has certain rights in the invention. INCORPORATION OF SEQUENCE LISTING
[0003] The sequence listing that is contained in the file named "UTFHP0293WO_ST25.txt", which is 8 KB (as measured in Microsoft Windows®) and was created on March 12, 2014, is filed herewith by electronic submission and is incorporated by reference herein. BACKGROUND OF THE INVENTION
1. Field of the Invention
[0004] The present invention relates generally to the field of cancer biology. More particularly, it concerns monoclonal antibodies targeting EpCAM antigen.
2. Description of Related Art
[0005] Dual-nuclear and fluorescently labeled or singly fluorescently labeled contrast agents promise the advantage of molecularly-guided surgical resection via surgical field near- infrared fluorescence (NIRF) imaging following (in the case of dual labeled agents) nuclear imaging for general localization. Currently, nodal staging of most cancers is performed following lymph node (LN) biopsy and dissection for subsequent pathological examination. As in most cancers, imaging of LN involvement in prostate cancer (PCa) lacks sensitivity and specificity (Shukla-Dave et ah, 2007) and as a result, (extended) pelvic LN dissection (PLND), which provides higher staging accuracy (Berney et ah, 201 1), is rapidly becoming the standard-of-care at the time of radical prostatectomy. Yet with early detection of PCa enabled by prostate-specific antigen (PSA) screening, a substantial and growing population of PCa patients may be overtreated (Daskivich et al, 2011) and encounter the resultant morbidity of lower extremity lymphedema (Pilepich et al, 1984; Musch et al, 2008; Cormier et al, 2010). A multimodal contrast agent is needed to provide accurate, non- invasive LN staging of PCa and to guide surgical resection of cancer-positive LNs, while sparing resection of cancer-negative LNs and potentially reducing surgical morbidity and improving survivorship.
[0006] In addition, surgical resection of epithelial cancers in body areas with critical areas, such as retroperitoneal or head and neck cancers require resection of tissues with clear margins free of cancer. Cancer positive margins, detected in surgical pathology from resected tissues, require surgeons to excise additional tissues, often times involving critical structures which compromise quality of life. Yet there is no indication within the field of surgical view, which structures are cancer positive and which are not. Because cancer survivorship critically depends upon reducing residual tumor burden by removal ALL cancerous tissues , it is critical that there is a means to direct surgical resection of cancerous tissues while sparing normal tissues, that may involve critical body structures.
SUMMARY OF THE INVENTION
[0007] The present invention provides methods and compositions for use in detecting and monitoring epithelial cancer cells. In some aspects, methods and compositions of the embodiments can be used for monitoring the spread of a epithelial cancer from primary tumor sites to the lymphatics and/or for identifying cancer-positive lymph nodes, or for delineating within the surgical field of view, cancer versus cancer free tissues.
[0008] In some embodiments, the present invention is directed towards an isolated or recombinant monoclonal antibody that specifically binds to a EpCAM polypeptide (e.g., a human EpCAM polypeptide). In certain aspects, an antibody competes for the binding of an EpCAM polypeptide with the "153" or "7" monoclonal antibody. In further aspects, an antibody of the embodiments can be defined as having an equilibrium ¾ relative to a human EpCAM polypeptide of less than about 5 nM or 10 nM, e.g., an equilibrium KD of between about 0.1 and 10 nM. In certain aspects, the antibody can comprise all or part of the heavy chain variable region and/or the light chain variable region of EpCAM-binding monoclonal antibodies. In a further aspect, a polypeptide can comprise an amino acid sequence that corresponds to a first, second, and/or third complementarity determining region (CDR) from the light variable and/or heavy variable chain of a EpCAM monoclonal antibody of the present invention.
[0009] Thus, in certain aspects, an isolated antibody comprises CDR sequences at least 80%, 90% or 95% identical to the CDR regions of the "153" or "7" antibody heavy and light chain amino acid sequences. In further aspects, an antibody comprises CDR regions identical to the 153 or 7, except for one or two amino acid substitutions, deletions or insertions at one or more of the CDRs. For example, the antibody can comprise CDRs wherein the CDR sequences comprise 1 or 2 amino acid substitutions in the VH CDRl, VH CDR2, VH CDR3, VL CDRl, VL CDR2 and/or VL CDR3 relative to the CDRs of a 153 or 7 monoclonal antibody. Thus, in some specific aspects, an antibody of the embodiments comprises (a) a first VH CDR at least 80% identical to VH CDRl of 153 (SEQ ID NO: 1), or 7 (SEQ ID NO: 9); (b) a second VH CDR at least 80% identical to VH CDR2 of 153 (SEQ ID NO: 2), or 7 (SEQ ID NO: 10); (c) a third VH CDR at least 80% identical to VH CDR3 of 153 (SEQ ID NO: 3), or 7 (SEQ ID NO: 1 1); (d) a first VL CDR at least 80% identical to VL CDRl of 153 (SEQ ID NO: 4), or 7 (SEQ ID NO: 12); (e) a second VL CDR at least 80% identical to VL CDR2 of 153 (SEQ ID NO: 5), or 7 (SEQ ID NO: 13); and (f) a third VL CDR at least 80% identical to VL CDR3 of 153 (SEQ ID NO: 6), or 7 (SEQ ID NO: 14).
[0010] In certain aspects, an antibody of the embodiments comprises each of the CDRs of a 153 antibody, such as, (a) a first VH CDR is identical to SEQ ID NO: 1; (b) a second VH CDR is identical to SEQ ID NO: 2; (c) a third VH CDR is identical to SEQ ID NO: 3; (d) a first VL CDR is identical to SEQ ID NO: 4; (e) a second VL CDR is identical to SEQ ID NO: 5; and (f) a third VL CDR is identical to SEQ ID NO: 6. Thus, in further aspects, the isolated antibody comprises a VH domain at least about 80% identical to the VH domain of 153 (SEQ ID NO: 7) and a VL domain at least about 80% identical to the VL domain of 153 (SEQ ID NO: 8). In some aspects, the isolated antibody comprises VH and VL domains identical to or derived from those of monoclonal antibody 153.
[0011] In certain aspects, an antibody of the embodiments comprises each of the CDRs of a 7 antibody, such as, (a) a first VH CDR is identical to SEQ ID NO: 9; (b) a second VH CDR is identical to SEQ ID NO: 10; (c) a third VH CDR is identical to SEQ ID NO: 1 1; (d) a first VL CDR is identical to SEQ ID NO: 12; (e) a second VL CDR is identical to SEQ ID NO: 13; and (f) a third VL CDR is identical to SEQ ID NO: 14. Thus, in further aspects, the isolated antibody comprises a VH domain at least about 80% identical to the VH domain of 7 (SEQ ID NO: 15) and a VL domain at least about 80% identical to the VL domain of 7 (SEQ ID NO: 16). In some aspects, the isolated antibody comprises VH and VL domains identical to or derived from those of monoclonal antibody 7.
[0012] In some aspects, an antibody of the embodiments may be an IgG (e.g., IgGl, IgG2, IgG3 or IgG4), IgM, IgA, or an antigen binding fragment thereof. The antibody may be a Fab', a F(ab')2 a F(ab')3, a monovalent scFv, a bivalent scFv, or a single domain antibody. The antibody may be a human, humanized, or de-immunized antibody. In a further aspect, the isolated antibody is the "153" or "7" monoclonal antibody and products derived thereof. [0013] In some aspects, an antibody may be conjugated to an imaging agent or a therapeutic agent. For example, the therapeutic agent can be a chemotherapeutic agent, a toxin, or a radionuclide (e.g., a radionuclide sequestered by a chelate). Imagining agents for use according to the embodiments include, without limitation, fluorophores, dyes, a MRI active agent, radionuclides or a fluorescence radionuclide. In certain aspects, an antibody can be conjugated to two or more agents, such as to a fluorophore and a radio-imaging agent.
[0014] In another embodiment, there is provided a recombinant polypeptide comprising an antibody VH domain comprising CDRs 1-3 of the VH domain of 153 (SEQ ID NOs: 1, 2 and 3); or CDRs 1-3 of the VH domain of 7 (SEQ ID NOs: 9, 10 and 1 1). In another embodiment a recombinant polypeptide is provided comprising an antibody VL domain comprising CDRs 1-3 of the VL domain of 153 (SEQ ID NOs: 4, 5 and 6); or 7 (SEQ ID NOs: 12, 13 and 14).
[0015] In some embodiments, there is provided an isolated polynucleotide molecule comprising nucleic acid sequence encoding an antibody or a polypeptide comprising an antibody VH or VL domain disclosed herein. In some aspects, the polynucleotide molecule is an expression vector.
[0016] In further embodiments, a host cell is provided that produces a monoclonal antibody or recombinant polypeptide of the embodiments. In some aspects, the host cell is a mammalian cell, a yeast cell, a bacterial cell, a ciliate cell, or an insect cell. In certain aspects, the host cell is a hybridoma cell. [0017] In still further embodiments, there is provided a method of manufacturing an antibody of the present embodiments comprising expressing one or more polynucleotide molecule(s) encoding a VL or VH chain of an antibody disclosed herein in a cell and purifying the antibody from the cell. [0018] In additional embodiments, pharmaceutical compositions are provided comprising an antibody or antibody fragment disclosed supra. Such a composition may further comprise a pharmaceutically acceptable carrier and may or may not contain additional active ingredients.
[0019] In yet further aspects, a polypeptide of the embodiments comprises an amino acid segment that is at least 80, 85, 90, 95, 96, 97, 98, 99, or 100% identical (or any range derivable therein) to a V, VJ, VDJ, D, DJ, J or CDR domain of a EpCAM-binding antibody (as provided in Table 1). For example, a polypeptide may comprise 1, 2 or 3 amino acid segment that are at least 80, 85, 90, 95, 96, 97, 98, 99, or 100% identical (or any range derivable therein) to CDRs 1, 2, and/or 3 a EpCAM-binding antibody as provided in Table 1. [0020] In still a further embodiment there is provided a method of preparing a subject for imaging, comprising administering to the subject an effective amount of an antibody of the embodiments, wherein the antibody is conjugated to an imaging agent. In a related embodiment a method is provided for imaging a subject comprising (a) administering an effective amount of an antibody of the embodiments to the subject, wherein the antibody is conjugated to an imaging agent; and (b) detecting the imaging agent in the subject. Depending on the imaging agent or agents used, detecting the imaging agent, can comprises performing fluorescence imaging, PET, MRI, CT, SPECT or a combination thereof on the subject. For example, in the case of NIR fluorescence imaging, images can be acquired using a fluorescence imaging system as described in U.S. Patents 7,328,059 and 8,084,753 and U.S. Patent Publications US20080064954, US20100305453, US201 10071403, US2011028081 1, as well as in Sevick-Muraca 2012 (Translation of near-infrared fluorescence imaging technologies: emerging clinical applications. Annu. Rev. Med. 63, 217-231), each of which is incorporated herein by reference. In a further aspect, the imaging agent can be a fluorophore, a dye, an MRI contrast agent or a radionuclide. In certain aspects, a subject for imaging is a subject having as cancer. Accordingly, in aspects, imaging a subject comprises imaging cancer cells in a subject. For example, a method can be defined as a method for detecting cancer metastasis, such as lymphoid metastasis (e.g., lymph node metastasis of a prostate cancer) or for delineating tumor margins and infiltrating cancer within the surgical field of view.
[0021] In another embodiment, there is provided a method for detecting a cancer in a subject comprising testing for the presence of elevated EpCAM relative to a control in a sample from the subject, wherein the testing comprises contacting the sample with an antibody disclosed herein. For example, the method may be an in vitro or in vivo method. Examples, of methods that can be used to test a sample for EpCAM expression include, without limitation, ELISA, immunohistochemistry (IHC), fluorescence microscopy, and flow cytometry. [0022] In embodiments of the present invention, there is provided a method for treating a subject having a cancer comprising administering an effective amount of an antibody disclosed herein. In certain aspects, the antibody is a monoclonal antibody of the present invention, such as 153 or 7 monoclonal antibody, or a recombinant polypeptide comprising antibody segments derived therefrom. In some aspects, the antibody is conjugated to a therapeutic agent, such as a toxin or chemotherapeutic agent. A method may further comprise administering at least a second anticancer therapy to the subject. Examples of the second anticancer therapy include, but are not limited to, surgical therapy, chemotherapy, radiation therapy, cryotherapy, hormonal therapy, immunotherapy, or cytokine therapy. Such a second therapy can be administered before, after or essentially simultaneously with an antibody therapeutic of the embodiments. In further aspects, the method may further comprise administering a composition of the present embodiments more than one time to the subject, such as, for example, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20 or more times.
[0023] In certain aspects, a cancer, as described herein, may be a breast cancer, lung cancer, head & neck cancer, prostate cancer, esophageal cancer, tracheal cancer, brain cancer, liver cancer, bladder cancer, stomach cancer, pancreatic cancer, ovarian cancer, uterine cancer, cervical cancer, testicular cancer, colon cancer, rectal cancer, skin cancer, or other epithelial cancer. In one aspect, the cancer is an EpCAM positive cancer, such as a positive prostate cancer. [0024] Antibody, therapeutics and imaging agents detailed herein may be administered systemically, locally or distally. In some aspects, an antibody may be administered intravenously, intradermally, intratumorally, intramuscularly, intraperitoneally or subcutaneously.
[0025] As used herein the specification, "a" or "an" may mean one or more. As used herein in the claim(s), when used in conjunction with the word "comprising", the words "a" or "an" may mean one or more than one.
[0026] The use of the term "or" in the claims is used to mean "and/or" unless explicitly indicated to refer to alternatives only or the alternatives are mutually exclusive, although the disclosure supports a definition that refers to only alternatives and "and/or." As used herein "another" may mean at least a second or more. [0027] Throughout this application, the term "about" is used to indicate that a value includes the inherent variation of error for the device, the method being employed to determine the value, or the variation that exists among the study subjects.
[0028] Other objects, features and advantages of the present invention will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples, while indicating preferred embodiments of the invention, are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description.
BRIEF DESCRIPTION OF THE DRAWINGS
[0029] The following drawings form part of the present specification and are included to further demonstrate certain aspects of the present invention. The invention may be better understood by reference to one or more of these drawings in combination with the detailed description of specific embodiments presented herein. [0030] FIG. 1. Dissociation analysis of mAbs from EpCAM/Fc by surface plasmon resonance (SPR) analysis. In series, the in-house panel of mAbs and commercial mAb 9601, were captured on a CM5 Sensor chip coupled with goat anti-mouse followed by EpCAM/Fc analyte. Dissociation of EpCAM/Fc from mAb is monitored as a decrease in response units (RU) over time. [0031] FIG. 2. Competitive binding analysis identified two independent epitopes recognized by the mAb panel. Unlabeled mAb clones (x-axis) bound EpCAM-coated ELISA plates followed by addition of either HRP-labeled anti-EpCAM mAb 9601 or 7. Competing and non-competing epitopes were visualized after washing and addition of HRP substrate. Competing epitopes of mAb 9601 (FIG. 2A) and mAb 7 (FIG. 2B) were those identified which inhibited binding of HRP-labeled mAb whereas non-competing epitopes did not inhibit binding of the labeled mAbs as monitored by HRP signaling (y-axis).
[0032] FIG. 3. Selected candidate mAb binding to native EpCAM expression on PC3 cells compared to that of commercial mAb 9601 via flow cytometry. Anti-EpCAM mAb 7, 153, or 9601 was incubated with PC3 cells followed by detection with PE-labeled secondary antibody. The geometric mean fluorescence intensity (MFI) of the cell population was determined by analysis of 10,000 cells using a BD FACSCalibur Flow Cytometer. The fold increase in MFI for cells incubated with each mAb is shown relative to the MFI of cells incubated with only PE-labeled secondary antibody. [0033] FIG. 4. Median fluorescence intensity (MFI) of PC3 cells bound by IRDye- labeled antibody (Ab), as determined by near-infrared (NIR) flow cytometry. After incubation with conjugated mAb 9601, 153, 7, or 69, or assay control Ab, PC3 cells were analyzed using a customized BD FACS Ariall. *P < 0.05 for mAbs 7 and 153 versus mAb 9601, and for mAb 7 versus mAb 153. **P < 0.05 for unlabeled mAb 9601 followed by IRDye-labeled secondary Ab versus IRDye-labeled secondary Ab alone. No significant difference (P > 0.05) was found for unlabeled mAb 9601 followed by IRDye-labeled secondary Ab versus conjugated mAb 9601.
[0034] FIG. 5. Non-invasive PET/CT and invasive DsRed and near-infrared (NIR) fluorescence images of a representative mouse having a primary prostate tumor and metastatic lymph nodes (LNs). Following dual-labeled mAb administration, coronal (FIG. 5A) and sagittal (FIG. 5B) views from PET/CT show radiotracer signal in the prostate region and several LNs. Regions of interest (ROI, white circles) were drawn around cancer-positive tissue using Inveon Research Workplace (IRW) software. The %ID/g was calculated for each ROI (including background muscle tissue) using IRW. The 3-D view from PET/CT (FIG. 5C) demonstrates better tissue delineation relative to single-slice 2-D views, including delineating primary tumor from bladder, metastatic renal LNs (RLNs) from kidneys and liver (where agent is cleared), and a metastatic sciatic LN (SLN). In situ DsRed fluorescence images (FIGs. 5D and G) of a ventral view from the same animal confirm the presence of a primary prostate tumor and cancer-positive lumbar LNs (LLNs) and RLNs via PC3 cell DsRed reporter-gene expression. The same cancer-positive tissues were detected via in situ NIR fluorescence imaging (FIGs. 5E and H) due to specific binding by dual-labeled mAb (agent). Anatomical location of tissue is depicted by corresponding white light photographs (FIGs. 5F and I) taken during in situ imaging.
[0035] FIG. 6. Receiver operating characteristic (ROC) curve analyses of TBR of %ID/g measured from PET/CT of LNs in mice given dual-labeled mAb 153, 7, 9601, or 69 (with ^Cu-DOTA and IRDye800). DsRed fluorescence from the gene reporter was considered the true-positive indicator of cancer-positive cells in LNs. The true-positive rate (TPR) or sensitivity was plotted versus false-positive rate (FPR or [1 - specificity]) for each mAb using ascending positive cut-off values, or criteria, that defined positive PET results. The ROC curve and area under the curve (AUC) for each mAb is demonstrated. While a perfect classification is defined as TPR = 1.0 and FPR = 0, mAbs 7 and 153 each demonstrated a significantly higher AUC or concordance rate (86.8% and 78.0%, respectively) relative to the 50% random chance line of no discrimination (dashed line). No significant differences were found between the concordance rates for mAbs 9601 or 69 relative to the 50% nondiscriminatory line. mAb 69 targets another molecule besides EpCAM and thus has imaging performance below the diagonal change line. [0036] FIG. 7. Analysis of mAb binding to human and mouse carcinoma cells. Anti-
EpCAM mAb 153 or G8.8 was incubated with either human MCF7 or mouse 4T1 cells. The mouse anti-human EpCAM mAb 153 and rat anti-mouse mAb G8.8 were detected with species-specific secondary antibodies, PE-labeled anti-mouse IgG, and DyLIGHT 488- labeled anti-rat IgG, respectively. The geometric mean fluorescence intensity (MFI) was determined with a BD FACSCalibur Flow Cytometer using WinMDI2.9 software. FIG. 7A demonstrates binding specificity of mAb 153 to human MCF7cells, as shown by the increase in MFI for the cell population incubated with mAb 153 (right- most peak) relative to the MFI for cells incubated with only PE-labeled secondary antibody (left-most peak), while binding of mAb 153 to mouse cells was not detectable (FIG. 7C), as shown by an MFI that was indistinguishable from that of the mouse cells incubated with only PE-labeled secondary antibody. FIGs. 7B and D demonstrate binding specificity of mAb G8.8 to mouse 4T1 cells and no detectable binding to the human cells. Additionally, the entire generated panel of anti-EpCAM mAbs were tested against 4T1 cells and, like mAb 153, demonstrated no detectable binding.
[0037] FIG. 8. DsRed fluorescence imaging video of animal LN staging. Longitudinal DsRed fluorescence imaging was performed in vivo at two-week intervals post- implantation of DsRed-expressing PC3 cells to monitor primary tumor growth and detect metastatic LNs if present. LN staging could be performed successfully in vivo for some mice, with or without holding the skin slightly taut. Metastases usually involved the lumbar LNs (as shown with holding), and sometimes medial iliac, renal, sciatic, inguinal, or popliteal LNs. [0038] FIG. 9. Comparison of in house generated mAbs 7 and 153 to Veridex mAb
VU-1D9 by flow cytometry. Using decreasing levels of primary antibody for labeling breast cancer cell line MCF7 followed by a fluorescently labeled secondary antibody, 10,000 cells from each population were analyzed by flow cytometry and MFI reported. Upon decreasing the level of primary antibody, in house mAbs 7 and 153 demonstrated improved ability to recognize and label surface expressed EpCAM on MCF7s. Numbers in parentheses represent MFI.
[0039] FIG. 10. Evaluation of scFv anti-EpCAM binding. Population of human prostate cancer cell line PC3 cells labeled with scFv 7 or scFv 153 detected with fluorophore conjugated secondary antibody and analyzed by flow cytometry (FIG. IOC), demonstrated mean fluorescent intensities (MFI) of 380 and 1164 compared to secondary control MFI of 10. Affinity of anti-EpCAM scFvs was determined by Biacore kinetic analysis. scFv 7 and scFv 153 were measured to have affinities of 4.14 nM and 0.21 nM, respectively (FIGs. 10A and B). This compares similarly to the affinity determination of Fab fragments produced via cleavage of parent mAbs 7 and 153, which demonstrated affinities of 2.3 nM and 0.54 nM, respectively. Slight differences may be attributed to accuracy in measuring active concentrations of Fab vs scFv prior to kinetic evaluation.
[0040] FIG. 11. Affinity maturation of lead antibody candidates using APEx directed evolution.
[0041] FIG. 12. Evaluation of population of 10,000 E. coli cells displaying anti- EpCAM scfv 7 and 153 via the APEx platform can be specifically labeled with EpCAM-Fc followed by detection with fluorescently conjugated secondary antibody with MFIs of 170 and 248, respectively. This is compared to an E. coli population expressing scFv with no affinity to EpCAM as a control, labeled under these conditions with a MFI of only 13. Affinity of scFv correlates with scFv affinity to EpCAM, with resolution dependent on stringency of the wash steps on E. coli prior to evaluation. DESCRIPTION OF ILLUSTRATIVE EMBODIMENTS
[0042] Epithelial cell adhesion molecule (EpCAM) is overexpressed in many metastasizing epithelial cancers (Spizzo et al, 2006; Spizzo et al, 2004; Went et al, 2006; Fong et al, 2008; Baeuerle et al, 2007) and is an ideal target for dual-labeled agent development. Expression of EpCAM in prostatic biopsy samples has been shown through immunohistochemistry (IHC) to be elevated with biochemical recurrence (Benko et al, 201 1) and correlated with Gleason score (Benko et al, 2011 ; Mukherjee et al, 2009), which is an important prognostic factor and is now incorporated into PCa staging nomograms (Edge et al, 2010). In over 441 human PCa tissue samples, microarray analyses showed that 80%- 100% of all prostatic intraepithelial neoplasia, localized PCa, hormone-refractory local recurrences, and lymph node (LN) metastases overexpressed EpCAM, suggesting it as an excellent diagnostic imaging marker for PCa LN staging (Zellweger et al, 2005).
[0043] Previously, the inventors developed a dual-labeled anti-EpCAM monoclonal antibody (mAb) for multimodal diagnostic imaging in a preclinical model of metastatic PCa, but optimization of binding affinity and pharmacodynamics of antibody (Ab)-based agents is needed to maximize imaging sensitivity and specificity (Hall et al, 2011). While anti- EpCAM mAbs have been under investigation as therapeutics against epithelial cancers, development has been focused upon attenuating binding affinity to reduce systemic toxicity, and enhancing Ab-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) for therapeutic action. As a diagnostic, the design of anti-EpCAM mAb- based imaging agents requires enhanced affinity for improved specificity at sub-therapeutic dosages, elimination of pharmacologic action through ADCC and CDC, and reduced circulation times to improve target-to-background ratios.
[0044] Herein, the inventors developed and quantitatively compared a panel of mAbs specific for human EpCAM, from which high-affinity candidates with specificity for non- overlapping EpCAM epitopes were dual-labeled with a near-infrared (NIR) fluorophore and a conventional chelator/radiotracer, for subsequent assessment via NIRF imaging and quantitative PET/CT for in vivo detection of metastatic LNs in a DsRed-reporter gene mouse model of PCa LN metastasis. The inventors sought to first assess whether differences in binding affinity impacted quantitative imaging performance of whole mAb-based contrast agents assessed by receiver operating characteristic (ROC) curve analyses prior to engineering mAb fragment-based imaging agents that will lack properties of therapeutic ADCC and CDC but will have improved imaging pharmacokinetics. A commercially available anti-EpCAM mAb, which was dual-labeled and used in previous preclinical imaging (Hall et al, 2011), was also included in the ROC curve analyses for comparison and selection of candidate mAbs for further imaging agent optimization.
[0045] Two high-affinity candidate mAbs with specificity for non-overlapping epitopes on the EpCAM surface antigen were chosen for further evaluation in vivo. After conjugation with 64Cu and IRDye800 CW, dual-labeled specific or isotype control mAb was administered intravenously to male Nu/Nu mice at 10-12 weeks post-orthotopic implantation of DsRed-expressing PC3 cells. Within 18-24 hours, non-invasive μΡΕΤ/CT was performed as well as in vivo and in situ DsRed reporter-gene and near-infrared fluorescence (NIRF) imaging to detect primary tumors and metastatic LNs. From quantitative analyses of noninvasive PET/CT images of LNs from the inventor's in vivo DsRed-reporter gene mouse model, the inventors demonstrated greater concordance between μΡΕΤ/CT imaging and the DsRed fluorescence, cancer-positive indicator using two in-house generated mAbs relative to the commercial mAb tested. MAbs 7 and 153 performed with greater sensitivity during μΡΕΤ/CT when compared to commercial mAb and yielded strong correlations between qualitative NIRF and DsRed-reporter gene imaging. Qualitative comparison of NIRF imaging demonstrated similar results to those found from DsRed reporter-gene imaging and μΡΕΤ/CT. MAbs 7 and 153 are attractive candidates for further agent optimization aimed at enhancing sensitivity and specificity for detection of metastatic LNs in PCa and creating Ab fragments to reduce circulation times and eliminate Fc binding regions responsible for therapeutic action and nonspecific binding.
[0046] There is a clear surgical need for improvements in cancer and metastasis detection. Although EpCAM has been a putative cancer therapeutic target for decades, this is one of the first efforts to specifically engineer the detection agent for diagnostic use. This could be used as an intraoperative means for surgeons to identify and specifically remove cancer positive lymph nodes during prostatectomy surgery and subsequent lymphadectomy. Additionally, for CTC detection, with the Veridex FDA approved platform, a clear association has been made with CTC quantification and lifespan of patient, demonstrating value of this technology in determining direction of patient treatment (Wallwiener et ah, 2013; Young et ah, 2012; Sun et ah, 2012). The next logical step is to improve the anti- EpCAM agent affinity as it is the primary means of CTC isolation. Improvements in affinity should translate into improved CTC detection while minimizing the blood sample volume necessary from the patient.
I. Definitions
[0047] As used herein, and unless otherwise indicated, the terms "treat," "treating," "treatment" and "therapy" contemplate an action that occurs while a patient is suffering from epithelial cancers and related disorders that reduces the severity of one or more symptoms or effects of epithelial cancers, metastisis or a related disease or disorder, such as but not limited to breast cancer, lung cancer, head & neck cancer, prostate cancer, esophageal cancer, tracheal cancer, brain cancer, liver cancer, bladder cancer, stomach cancer, pancreatic cancer, ovarian cancer, uterine cancer, cervical cancer, testicular cancer, colon cancer, rectal cancer, skin cancer, or other epithelial cancer and metastasis associated with such. In one aspect, the cancer is an EpCAM positive cancer, such as a positive prostate cancer.
[0048] Where the context allows, the terms "treat," "treating," and "treatment" also refers to actions taken toward ensuring that individuals at increased risk of epithelial cancer - metastasis are able to receive appropriate surgical and/or other medical intervention prior to onset of epithelial cancers and disorders. As used herein, and unless otherwise indicated, the terms "prevent," "preventing," and "prevention" contemplate an action that occurs before a patient begins to suffer from epithelial cancers and related disorders that delays the onset of, and/or inhibits or reduces the severity of, epithelial cancers and related disorders. As used herein, and unless otherwise indicated, the terms "manage," "managing," and "management" encompass preventing, delaying, or reducing the severity of a recurrence of epithelial cancers, metastisis and related disorders in a patient who has already suffered from such a disease or condition. The terms encompass modulating the threshold, development, and/or duration of the epithelial cancers, metastisis and related disorders or changing how a patient responds to the epithelial cancers, metastisis and related disorders. [0049] As used herein, and unless otherwise specified, a "therapeutically effective amount" of a compound is an amount sufficient to provide any therapeutic benefit in the treatment or management of epithelial cancers, metastisis and related disorders or to delay or minimize one or more symptoms associated with epithelial cancers, metastisis and related disorders. A therapeutically effective amount of a compound means an amount of the compound, alone or in combination with one or more other therapies and/or therapeutic agents, which provides any therapeutic benefit in the treatment or management of epithelial cancers, metastisis and related disorders, or related diseases or disorders. The term "therapeutically effective amount" can encompass an amount that alleviates epithelial cancers, metastisis and related disorders, improves or reduces epithelial cancers, metastisis and related disorders, improves overall therapy, or enhances the therapeutic efficacy of another therapeutic agent. The "therapeutically effective amount" can be identified at an earlier stage with parameters of 1 epithelial cancers, metastisis and related epithelial cancers, metastisis and related function as identified herein.
[0050] As used herein, the term "lymphatic structure(s)" refers to all or a portion of structures that make up a mammalian lymphatic system including without limitation, lymph nodes, collecting vessels, lymph trunks, lymph ducts, capillaries, or combinations thereof. The architecture of the lymphatic structures can be described by tortuosity, density, dilation, and other parameters.
[0051] As used herein, the term "near-infrared" refers to electromagnetic radiation at wavelengths ranging from about 750 nm to about 900 nm.
[0052] As used herein, and unless otherwise specified, the term "functional imaging" of lymph structures refers to how the structures function in terms of update of dye, the lymphatic flow as determined by the dye, dynamics of flow, and direction of flow of lymph and the associated materials carried by it. The function of the lymphatic structures can be described by lymph velocity, period or frequency of propulsive events, permeability, and other parameters that provide evidence of dysfunction in comparison to normal function imaged in healthy control animals or human subjects. If lymph vessels are "functional" they transport materials, and imaging methods disclosed herein describe this.
II. EpCAM
[0053] EpCAM, a 40 kD type I transmembrane glycoprotein involved in cell-to-cell interactions and adhesion, is stably overexpressed by most types of epithelial cancers. EpCAM was the first identified tumor-associated antigen that today is known to be intensely and uniformly expressed on epithelial carcinomas while being less intensely expressed, sequestered, and less accessible on the basolateral cell surface of normal epithelia, making it a useful marker for diagnostic applications.
[0054] Proliferation of most carcinomas is associated with overexpression of epithelial cell adhesion molecule (EpCAM), a 40 kDa Type I transmembrane protein found on all epithelial cells yet absent on other cell types, including those in lymphoid tissue. Absence of EpCAM in normal lymphatics makes it an attractive marker for studying lymph node (LN) metastases of carcinomas to improve LN staging accuracy.
[0055] These monoclonal antibodies have demonstrated higher affinity and specificity compared to commercially available anti-EpCAM antibody 9601 (R&D Systems). III. Diagnostic Antibodies
[0056] In certain embodiments, an antibody or a fragment thereof that binds to at least a portion of EpCAM protein and its associated use in diagnosis and imaging of diseases are contemplated. As used herein, the term "antibody" is intended to refer broadly to any immunologic binding agent, such as IgG, IgM, IgA, IgD, and IgE as well as polypeptides comprising antibody CDR domains that retain antigen binding activity. The antibody may be selected from the group consisting of a chimeric antibody, an affinity matured antibody, a polyclonal antibody, a monoclonal antibody, a humanized antibody, a human antibody, or an antigen-binding antibody fragment or a natural or synthetic ligand. Preferably, the anti- EpCAM antibody is a monoclonal antibody or a humanized antibody. By known means and as described herein, polyclonal or monoclonal antibodies, antibody fragments, and binding domains and CDRs (including engineered forms of any of the foregoing) may be created that are specific to EpCAM protein, one or more of its respective epitopes, or conjugates of any of the foregoing, whether such antigens or epitopes are isolated from natural sources or are synthetic derivatives or variants of the natural compounds. [0057] Examples of antibody fragments suitable for the present invention include, without limitation: (i) the Fab fragment, consisting of VL, VH, CL, and CHI domains; (ii) the "Fd" fragment consisting of the VH and CHI domains; (iii) the "Fv" fragment consisting of the VL and VH domains of a single antibody; (iv) the "dAb" fragment, which consists of a VH domain; (v) isolated CDR regions; (vi) F(ab')2 fragments, a bivalent fragment comprising two linked Fab fragments; (vii) single chain Fv molecules ("scFv"), wherein a VH domain and a VL domain are linked by a peptide linker that allows the two domains to associate to form a binding domain; (viii) bi-specific single chain Fv dimers (see U.S. Pat. No. 5,091,513); and (ix) diabodies, multivalent or multispecific fragments constructed by gene fusion (US Patent App. Pub. 20050214860). Fv, scFv, or diabody molecules may be stabilized by the incorporation of disulphide bridges linking the VH and VL domains. Minibodies comprising a scFv joined to a CH3 domain may also be made (Hu et ah, 1996).
[0058] Antibody-like binding peptidomimetics are also contemplated in embodiments. Liu et al. (2003) describe "antibody like binding peptidomimetics" (ABiPs), which are peptides that act as pared-down antibodies and have certain advantages of longer serum half-life as well as less cumbersome synthesis methods. [0059] Animals may be inoculated with an antigen, such as purified peptide or cells expressing the antigen on their surface, in order to produce antibodies specific for EpCAM protein. Frequently an antigen is bound or conjugated to another molecule to enhance the immune response. As used herein, a conjugate is any peptide, polypeptide, protein, or non- proteinaceous substance bound to an antigen that is used to elicit an immune response in an animal. Antibodies produced in an animal in response to antigen inoculation comprise a variety of non-identical molecules (polyclonal antibodies) made from a variety of individual antibody producing B lymphocytes. A polyclonal antibody is a mixed population of antibody species, each of which may recognize a different epitope on the same antigen. Given the correct conditions for polyclonal antibody production in an animal, most of the antibodies in the animal's serum will recognize the collective epitopes on the antigenic compound to which the animal has been immunized. This specificity is further enhanced by affinity purification to select only those antibodies that recognize the antigen or epitope of interest.
[0060] A monoclonal antibody is a single species of antibody wherein every antibody molecule recognizes the same epitope because all antibody producing cells are derived from a single B-lymphocyte cell line. The methods for generating monoclonal antibodies (MAbs) generally begin along the same lines as those for preparing polyclonal antibodies. In some embodiments, rodents such as mice and rats are used in generating monoclonal antibodies. In some embodiments, rabbit, sheep, or frog cells are used in generating monoclonal antibodies. The use of rats is well known and may provide certain advantages. Mice (e.g., BALB/c mice) are routinely used and generally give a high percentage of stable fusions. [0061] Hybridoma technology involves the fusion of a single B lymphocyte from a mouse previously immunized with a EpCAM antigen with an immortal myeloma cell (usually mouse myeloma). This technology provides a method to propagate a single antibody-producing cell for an indefinite number of generations, such that unlimited quantities of structurally identical antibodies having the same antigen or epitope specificity (monoclonal antibodies) may be produced.
[0062] In one embodiment, the antibody is a chimeric antibody, for example, an antibody comprising antigen binding sequences from a non-human donor grafted to a heterologous non-human, human, or humanized sequence (e.g., framework and/or constant domain sequences). Methods have been developed to replace light and heavy chain constant domains of the monoclonal antibody with analogous domains of human origin, leaving the variable regions of the foreign antibody intact. Alternatively, "fully human" monoclonal antibodies are produced in mice transgenic for human immunoglobulin genes. Methods have also been developed to convert variable domains of monoclonal antibodies to more human form by recombinantly constructing antibody variable domains having both rodent, for example, mouse, and human amino acid sequences. In "humanized" monoclonal antibodies, only the hypervariable CDR is derived from mouse monoclonal antibodies, and the framework and constant regions are derived from human amino acid sequences (see U.S. Pat. Nos. 5,091,513 and 6,881,557). It is thought that replacing amino acid sequences in the antibody that are characteristic of rodents with amino acid sequences found in the corresponding position of human antibodies will reduce the likelihood of adverse immune reaction during therapeutic use. A hybridoma or other cell producing an antibody may also be subject to genetic mutation or other changes, which may or may not alter the binding specificity of antibodies produced by the hybridoma. [0063] Methods for producing polyclonal antibodies in various animal species, as well as for producing monoclonal antibodies of various types, including humanized, chimeric, and fully human, are well known in the art and highly predictable. For example, the following U.S. patents and patent applications provide enabling descriptions of such methods: U.S. Patent Application Nos. 2004/0126828 and 2002/0172677; and U.S. Pat. Nos. 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4, 196,265; 4,275, 149; 4,277,437; 4,366,241; 4,469,797; 4,472,509; 4,606,855; 4,703,003; 4,742,159; 4,767,720; 4,816,567; 4,867,973; 4,938,948; 4,946,778; 5,021,236; 5, 164,296; 5, 196,066; 5,223,409; 5,403,484; 5,420,253; 5,565,332; 5,571,698; 5,627,052; 5,656,434; 5,770,376; 5,789,208; 5,821,337; 5,844,091; 5,858,657; 5,861, 155; 5,871,907; 5,969, 108; 6,054,297; 6, 165,464; 6,365, 157; 6,406,867; 6,709,659; 6,709,873; 6,753,407; 6,814,965; 6,849,259; 6,861,572; 6,875,434; and 6,891,024. All patents, patent application publications, and other publications cited herein and therein are hereby incorporated by reference in the present application.
[0064] Antibodies may be produced from any animal source, including birds and mammals. Preferably, the antibodies are ovine, murine (e.g., mouse and rat), rabbit, goat, guinea pig, camel, horse, or chicken. In addition, newer technology permits the development of and screening for human antibodies from human combinatorial antibody libraries. For example, bacteriophage antibody expression technology allows specific antibodies to be produced in the absence of animal immunization, as described in U.S. Pat. No. 6,946,546, which is incorporated herein by reference. These techniques are further described in: Marks (1992); Stemmer (1994); Gram et al. (1992); Barbas et al. (1994); and Schier et al. (1996).
[0065] It is fully expected that antibodies to EpCAM will have the ability to bind EpCAM regardless of the animal species, monoclonal cell line, or other source of the antibody. Certain animal species may be less preferable for generating diagnostic imaging antibodies because they may be more likely to cause allergic response due to activation of the complement system through the "Fc" portion of the antibody. However, whole antibodies may be enzymatically digested into "Fc" (complement binding) fragment, and into antibody fragments having the binding domain or CDR. Removal of the Fc portion reduces the likelihood that the antigen antibody fragment will elicit an undesirable immunological response, and thus, antibodies without Fc may be preferential for prophylactic or therapeutic treatments. As described above, antibodies may also be constructed so as to be chimeric or partially or fully human, so as to reduce or eliminate the adverse immunological consequences resulting from administering to an animal an antibody that has been produced in, or has sequences from, other species.
[0066] Substitutional variants typically contain the exchange of one amino acid for another at one or more sites within the protein, and may be designed to modulate one or more properties of the polypeptide, with or without the loss of other functions or properties. Substitutions may be conservative, that is, one amino acid is replaced with one of similar shape and charge. Conservative substitutions are well known in the art and include, for example, the changes of: alanine to serine; arginine to lysine; asparagine to glutamine or histidine; aspartate to glutamate; cysteine to serine; glutamine to asparagine; glutamate to aspartate; glycine to proline; histidine to asparagine or glutamine; isoleucine to leucine or valine; leucine to valine or isoleucine; lysine to arginine; methionine to leucine or isoleucine; phenylalanine to tyrosine, leucine or methionine; serine to threonine; threonine to serine; tryptophan to tyrosine; tyrosine to tryptophan or phenylalanine; and valine to isoleucine or leucine. Alternatively, substitutions may be non-conservative such that a function or activity of the polypeptide is affected. Non-conservative changes typically involve substituting a residue with one that is chemically dissimilar, such as a polar or charged amino acid for a nonpolar or uncharged amino acid, and vice versa. [0067] Proteins may be recombinant, or synthesized in vitro. Alternatively, a non- recombinant or recombinant protein may be isolated from bacteria. It is also contemplated that a bacteria containing such a variant may be implemented in compositions and methods. Consequently, a protein need not be isolated.
[0068] An antibody or preferably an immunological portion of an antibody, can be chemically conjugated to, or expressed as, a fusion protein with other proteins. For purposes of this specification and the accompanying claims, all such fused proteins are included in the definition of antibodies or an immunological portion of an antibody.
[0069] Embodiments provide antibodies and antibody-like molecules against EpCAM, polypeptides and peptides that are linked to at least one agent to form an antibody conjugate or payload. In order to increase the efficacy of antibody molecules as diagnostic or therapeutic agents, it is conventional to link or covalently bind or complex at least one desired molecule or moiety. Such a molecule or moiety may be, but is not limited to, at least one effector or reporter molecule. Effector molecules comprise molecules having a desired activity, e.g., cytotoxic activity. Non-limiting examples of effector molecules that have been attached to antibodies include toxins, therapeutic enzymes, antibiotics, radio-labeled nucleotides and the like. By contrast, a reporter molecule is defined as any moiety that may be detected using an assay. Non-limiting examples of reporter molecules that have been conjugated to antibodies include enzymes, radiolabels, haptens, fluorescent labels, phosphorescent molecules, chemiluminescent molecules, chromophores, luminescent molecules, photoaffinity molecules, colored particles or ligands, such as biotin. [0070] Several methods are known in the art for the attachment or conjugation of an antibody to its conjugate moiety. Some attachment methods involve the use of a metal chelate complex employing, for example, an organic chelating agent such a diethylenetriaminepentaacetic acid anhydride (DTPA); ethylenetriaminetetraacetic acid; N- chloro-p-toluenesulfonamide; and/or tetrachloro-3-6?-diphenylglycouril-3 attached to the antibody. Monoclonal antibodies may also be reacted with an enzyme in the presence of a coupling agent such as glutaraldehyde or periodate. Conjugates with fluorescein markers are prepared in the presence of these coupling agents or by reaction with an isothiocyanate.
[0071] The conjugation of the radionuclide to the antibodies or fragments of the invention can be made using any conventional techniques such as the use of a linker between the antibody and the radioisotope. Preferably, the radioimmunoconjugate has a specific activity of from about 0.5 to about 15 mCi/mg, depending on the radionuclide, and may be administered via an intravenous or other route.
IV. Diagnostic Imaging of Diseases
[0072] Certain aspects of the present invention can be used to diagnose and/or image a disease or disorder associated with EpCAM expression, for example, prostate cancer. In this regard, an antibody or antibody fragment can be detectably labeled through the use of radioisotopes, affinity labels (e.g., biotin, avidin, etc.), fluorescent labels, paramagnetic atoms, etc. The detection of foci of such detectably labeled antibodies or antibody fragments might be indicative of a site of tumor development. In a preferred embodiment, this technique is done in a non-invasive manner through the use of magnetic imaging, fluorography, PET, etc. Such a diagnostic test may be employed in monitoring the success of treatment of diseases, where presence or absence of EpCAM-positive cells is a relevant indicator. Such imaging may also be employed during surgical resection of lymph nodes comprising metastatic disease to guide said surgical resection.
[0073] Visualization and quantification of MAb biodistribution using PET requires a suitable positron-emitting radionuclide. 89Zr (t1/2 = 78.4 h), 124I (t1/2 = 100.3 h), 64Cu (t1/2 = 12.7 h), 86Y (ti/2 = 14.7 h), and 76Br (t1/2 = 16.2 h) are well suited for imaging of intact MAbs.
[0074] Where clinical application of a diagnostic composition containing an inhibitory antibody is undertaken, it will generally be beneficial to prepare a pharmaceutical composition appropriate for the intended application. In certain embodiments, pharmaceutical compositions may comprise, for example, at least about 0.1% of an active compound. In other embodiments, an active compound may comprise between about 2% to about 75% of the weight of the unit, or between about 25% to about 60%, for example, and any range derivable therein. [0075] The pharmaceutical compositions of the present invention are advantageously administered in the form of injectable compositions either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid prior to injection may also be prepared. These preparations also may be emulsified.
[0076] The phrases "pharmaceutical or pharmacologically acceptable" refers to molecular entities and compositions that do not produce an adverse, allergic, or other untoward reaction when administered to an animal, such as a human, as appropriate. The preparation of a pharmaceutical composition comprising an antibody or additional active ingredient will be known to those of skill in the art in light of the present disclosure. Moreover, for animal (e.g., human) administration, it will be understood that preparations should meet sterility, pyrogenicity, general safety, and purity standards as required by FDA Office of Biological Standards.
[0077] As used herein, "pharmaceutically acceptable carrier" includes any and all aqueous solvents (e.g., water, alcoholic/aqueous solutions, saline solutions, parenteral vehicles, such as sodium chloride, Ringer's dextrose, etc.), non-aqueous solvents (e.g., propylene glycol, polyethylene glycol, vegetable oil, and injectable organic esters, such as ethyloleate), dispersion media, coatings, surfactants, antioxidants, preservatives (e.g., antibacterial or antifungal agents, anti-oxidants, chelating agents, and inert gases), isotonic agents, absorption delaying agents, salts, drugs, drug stabilizers, gels, binders, excipients, disintegration agents, lubricants, sweetening agents, flavoring agents, dyes, fluid and nutrient replenishers, such like materials and combinations thereof, as would be known to one of ordinary skill in the art. The pH and exact concentration of the various components in a pharmaceutical composition are adjusted according to well-known parameters. A pharmaceutically acceptable carrier is particularly formulated for administration to a human, although in certain embodiments it may be desirable to use a pharmaceutically acceptable carrier that is formulated for administration to a non-human animal but that would not be acceptable (e.g., due to governmental regulations) for administration to a human. Except insofar as any conventional carrier is incompatible with the active ingredient, its use in the therapeutic or pharmaceutical compositions is contemplated.
[0078] The term "unit dose" or "dosage" refers to physically discrete units suitable for use in a subject, each unit containing a predetermined quantity of the diagnositc composition calculated to produce the desired responses discussed above in association with its administration, i.e., the appropriate route and treatment regimen. The quantity to be administered, both according to number of treatments and unit dose, depends on the effect desired. The actual dosage amount of a composition of the present invention administered to a patient or subject can be determined by physical and physiological factors, such as body weight, the age, health, and sex of the subject, the type of disease being imaged, the extent of disease penetration, previous or concurrent therapeutic interventions, idiopathy of the patient, the route of administration, and the potency, stability, and toxicity of the particular therapeutic substance. For example, a dose may also comprise from about 1 μg/kg/body weight to about 1000 mg/kg/body weight (this such range includes intervening doses) or more per administration, and any range derivable therein. In non-limiting examples of a derivable range from the numbers listed herein, a range of about 5 μg/kg/body weight to about 100 mg/kg/body weight, about 5 μg/kg/body weight to about 500 mg/kg/body weight, etc., can be administered. The practitioner responsible for administration will, in any event, determine the concentration of active ingredient(s) in a composition and appropriate dose(s) for the individual subject.
[0079] The active compounds can be formulated for parenteral administration, e.g., formulated for injection via the intravenous, intramuscular, sub-cutaneous, or even intraperitoneal routes. Typically, such compositions can be prepared as either liquid solutions or suspensions; solid forms suitable for use to prepare solutions or suspensions upon the addition of a liquid prior to injection can also be prepared; and, the preparations can also be emulsified.
[0080] The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil, or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases the form must be sterile and must be fluid to the extent that it may be easily injected. It also should be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
[0081] The proteinaceous compositions may be formulated into a neutral or salt form. Pharmaceutically acceptable salts, include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like. [0082] A pharmaceutical composition can include a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion, and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
V. Kits
[0083] In various aspects of the invention, a kit is envisioned containing diagnostic agents and/or other therapeutic and delivery agents. In some embodiments, the present invention contemplates a kit for preparing and/or administering a diagnostic agent of the invention. The kit may comprise one or more sealed vials containing any of the pharmaceutical compositions of the present invention. The kit may include, for example, at least one EpCAM antibody as well as reagents to prepare, formulate, and/or administer the components of the invention or perform one or more steps of the inventive methods. In some embodiments, the kit may also comprise a suitable container, which is a container that will not react with components of the kit, such as an eppendorf tube, an assay plate, a syringe, a bottle, or a tube. The container may be made from sterilizable materials such as plastic or glass. [0084] The kit may further include an instruction sheet that outlines the procedural steps of the methods set forth herein, and will follow substantially the same procedures as described herein or are known to those of ordinary skill in the art. The instruction information may be in a computer readable media containing machine-readable instructions that, when executed using a computer, cause the display of a real or virtual procedure of delivering a pharmaceutically effective amount of a diagnostic agent.
VI. Examples
[0085] The following examples are included to demonstrate preferred embodiments of the invention. It should be appreciated by those of skill in the art that the techniques disclosed in the examples which follow represent techniques discovered by the inventor to function well in the practice of the invention, and thus can be considered to constitute preferred modes for its practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention.
Example 1 - Materials and Methods
[0086] Monoclonal Antibody Panel Generation. Commercially available mouse IgG2b monoclonal Ab (mAb) 9601, specific for human EpCAM, was purchased from R & D Systems (Minneapolis, MN) and maintained at -20°C prior to labeling. To generate in-house anti-EpCAM mAbs, two separate immunization strategies were pursued: BALB/c mice were immunized with either recombinant human (rhu) EpCAM/Fc (960-EP, R&D Systems, Minneapolis, MN) or MCF7 cells (HTB-22™, ATCC, Manassas, VA), a human breast adenocarcinoma line that expresses EpCAM (Prang et ah, 2005; Munz et ah, 2010). EpCAM /Fc was administered using standard methods as previously described (Pinkston et ah, 2011). For whole-cell immunization, MCF7 cells were grown in GIBCO® Dulbecco's Minimal Essential Medium (DMEM) (Invitrogen Corporation, Carlsbad, CA) with 10% fetal bovine serum (FBS) to 80%-100% confluency in T75 flasks, detached with Trypsin- ethylenediaminetetraacetic acid (EDTA) (Cellgro, Manassas, VA), washed in phosphate buffered saline (PBS), pH 7.4, and injected intraperitoneally (ip) at 107 cells per mouse every ten days for five injections. Mouse splenocytes were collected and fused with SP2/0 mouse myeloma cells (Pinkston et ah, 2011). MAbs from parental wells were evaluated for binding specificity to rhuEpCAM/Fc via an enzyme-linked immunosorbent assay (ELISA) followed by kinetic screening to rank order high affinity clones using surface plasmon resonance (SPR) (Canziani et al, 2004).
[0087] Competitive Binding Assays. To compare relative binding sites of the inventor's mAb panel, a competitive binding strategy was used to separate the inventor's panel of mAbs into groups with competing and non-competing epitope binding sites. Briefly, a horse-radish peroxidase (HRP) labeling kit (Zymed Laboratories, San Francisco, CA) was used to HRP-label in-house generated mAbs or commercial mAb 9601. RhuEpCAM/Fc was coated on Microlon® 600 (Greiner Bio-One, Frickenhausen, Germany), 96-well, high- binding plates in PBS at 0.5 μg/ml and 4°C overnight. After blocking with PBS containing 0.02% Tween-20 and 5% Non-Fat Dry Milk for 1 h and a subsequent wash with PBS, 0.02% Tween (PBST), each selected mAb from the inventor's panel was added to the plate at 2 μg/ml and incubated for 30 min at room temperature (RT). This was followed by the addition of HRP -conjugated mAb at 0.2 μg/ml and incubation for 30 min at RT. Ability of HRP-conjugated mAb to bind rhuEpCAM/Fc was detected via reaction with 3, 3', 5,5'- tetramethylbenzidine (TMB) substrate (Sigma-Aldrich, St. Louis, MO) and measurement of OD 450 nm, using the manufacturer's protocol. Binding of HRP-labeled mAb demonstrated that epitopes were available for binding, while absence of binding by HRP-labeled mAb indicated competitive epitope binding by the primary mAb.
[0088] Preparation of Fab Fragments. Purified mouse IgGi, in-house mAbs 7 and 153, were digested with immobilized Ficin, Kit 44980 (Pierce, Rockford, IL), according to the manufacture's protocol. Commercial mAb 9601, IgG2b, was digested with immobilized Papain, Kit 44985 (Pierce). Briefly, purified IgG was dialyzed overnight and buffer exchanged with provided digestion buffer(s) without Cysteine-HCl. After a digestion period of 3-4 h, samples were applied to the provided protein A column. The pass-through material containing fragment antigen-binding (Fab) regions was dialyzed against PBS and retained for enrichment via size exclusion chromatography. A GE ΑΚΤΑ Explorer (GE Healthcare, Piscataway, NJ) fast protein liquid chromatography (FPLC) system coupled with a Superdex 200 HR 10/30 FPLC column (GE Healthcare) pre-equilibrated with PBS was utilized to separate the Fab from undigested IgG and F(ab')2. Purity was then assessed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE).
[0089] Surface Plasmon Resonance Assessment of Affinity Constant, KD. In order to discriminate between monovalent and multivalent interactions on quantitative SPR measurements of KD, the binding affinities of the Fab fragments of mAbs were measured using SPR and a Biacore T100 instrument (GE Healthcare). Two flow cells of a CM5 chip were coated with goat anti-human IgG (Fc-specific) (Jackson ImmunoResearch) to approximately 8000 resonance units (RU) with an NHS/EDC Amine Coupling Kit BR- 1000- 50 (GE Healthcare). All measurements were made at 25°C using HBS-EP+ (10 mM HEPES, 150 mM NaCl, 3 mM EDTA, 0.05% v/v Tween 20, pH 7.4) as running buffer. EpCAM/Fc ligand (R&D Systems) was diluted in HBS-EP+ to 0.5 μg/ml and applied to the coated chip surface. Fab fragments were then tested at five different molar concentrations starting at 50 nM after performing a two-fold dilution series. Flow cells were regenerated with 100 mM phosphoric acid. All samples were applied in duplicate.
[0090] Confirmation of MAb Whole-Cell Binding Activity. PC3 cells (human prostate adenocarcinoma line, CRL-1435™, ATCC, Manassas, VA), grown to 80%- 100% confluency in DMEM with 10% FBS, were treated with Trypsin-EDTA and resuspended in PBS containing 2% bovine serum albumin (BSA). One million cells per well were labeled with 5 μg/ml of mAb in a 200-μ1 volume and incubated on ice for 20 min. After washing with PBS-BSA, phycoerythrin (PE)-conjugated donkey F(ab')2 anti-mouse IgG (Jackson ImmunoResearch, West Grove, PA) at 5 μg per sample was added for an additional 20-min incubation. Cells were washed again with PBS-BSA and fixed in 2% paraformaldehyde prior to analyzing (10,000 events) on a BD FACSCalibur Flow Cytometer (BD Biosciences, San Jose, CA). Analyses of mean fluorescence intensities (MFI) were performed using WinMDI2.9 software.
[0091] Production of MAbs for Animal Model. One-liter cultures of hybridomas producing selected mAbs were grown in GIBCO® DMEM high glucose media (Invitrogen Corp.) supplemented with 2% FBS for 10 days. Supernatants were centrifuged, filtered (0.2 μιη), and purified on a Protein G column (GE Healthcare) per the manufacturer's instructions. Samples were dialyzed against PBS, filtered (0.2 μιη), and stored at 4°C until use.
[0092] Testing Species Cross-reactivity of MAbs. To determine whether mAbs specific for human EpCAM cross-react with murine EpCAM, the human cell line MCF7 (as described above) and 4T1 cells (mouse mammary carcinoma, CRL-2539™, ATCC) were used. Hybridoma cells were grown and mAbs purified as above-described. Rat anti-mouse EpCAM, mAb G8.8 (Developmental Studies Hybridoma Bank, University of Iowa, Iowa City, IA) was utilized as a positive control for detection of EpCAM on the surface of 4T 1 cells. Following incubation (20 min, on ice) with mAb, cells were washed with PBS-BSA and either DyLIGHT-488 conjugated donkey anti-rat IgG (Jackson ImmunoResearch) or PE- conjugated donkey F(ab') 2 anti-mouse IgG (Jackson ImmunoResearch) was added at 5 μg per sample and incubated for an additional 20 min for detecting cells bound by rat anti-mouse or mouse anti-human EpCAM mAb, respectively. Cells were washed again with PBS-BSA and fixed in 2% paraformaldehyde prior to analyzing (10,000 events) on a BD FACSCalibur Flow Cytometer (BD Biosciences) using WinMDI2.9 software.
[0093] Preparation of Dual-Labeled MAbs. MAbs were dual-labeled with DOTA chelator and IRDye 800CW, purified, and the number of chelates and NIR dye molecules conjugated per mAb was determined according to procedures described previously (Hall et ah, 201 1 ; Meares et ah, 1984). Radiolabeling of mAb-conjugates was performed by adding 37 MBq (1 mCi) of 64CuC12 (Radiological Sciences, Washington University Medical School, St. Louis, MO) to 100 μg of mAb-conjugate in 0.1 M NaOAc at pH 6.0, followed by incubation at 40°C for 1 h. The reactions were purified with spin columns as described above and analyzed by radio-thin layer chromatography (TLC) and radio-high-performance liquid chromatography (HPLC).
[0094] Determination of Contrast Agent Biological Activity. NIR flow cytometry was utilized to measure biological activity of the mAb-conjugates prior to use in animals. Human PCa cells (PC3 cell line, ATCC, Manassas, VA), which overexpress EpCAM (Poczatek et ah, 1999), were incubated (1 x 106) with fluorophore-labeled anti-EpCAM specific or isotype control mAb (1 μg) in 200 μΐ PBS, pH 7.4, at 37°C with 5% C02 for 50 min in the dark. A commercial IgG2b isotype control mAb (BD Pharmingen, San Diego, CA) and an in-house generated IgGi isotype control mAb were used in each assay to test for nonspecific binding. After centrifugation at 350 x g for 5 min at RT, supernatants were discarded and cells were washed with PBS. As a positive control, IRDye-labeled secondary Ab, goat anti-mouse IgG (LI-COR), was then added to reactions containing cells incubated with unlabeled, commercial mAb 9601. As a negative control for secondary Ab staining, the secondary Ab was also added to cells without primary Ab. Following incubation for 40 min with secondary Ab or PBS, cells were centrifuged as above-described, resuspended in 500 μΐ PBS, and placed on ice. Analysis immediately followed using a customized BD FACSAria™ II with NIR wavelength excitation and emission detection capability (BD Biosciences San Diego, CA). The percentage (%) of cells bound or stained by fluorophore-labeled Ab as well as the median fluorescence intensity (MFI) of the cells in each reaction was recorded. The means and standard deviations (SD) were calculated from three experiments for each mAb tested. [0095] Animal Model. A DsRed gene-reporter mouse model of PCa LN metastasis was utilized as previously described (17). In brief, 6-8 week-old, male Nu/Nu mice (Charles River, Wilmington, MA) were housed and maintained in compliance with protocols approved by the Animal Welfare Committee at the University of Texas Health Science Center at Houston, and with the Association for Assessment and Accreditation of Laboratory Animal Care. PC3 cells (ATCC), which were transfected with p-DsRedExpress-Nl (Clontech Laboratories, Inc., Mountain View, CA) and sorted as previously described (Hall et ah, 201 1), were orthotopically implanted (1 x 106 cells) in the dorsal prostate of each mouse. Non-invasive, longitudinal DsRed fluorescence imaging was performed bi-weekly to monitor tumor growth. [0096] Dual-labeled mAbs (40-50 μg/7.4-12.95 MBq) were administered via the tail vein at 10-12 weeks post-implantation. Within 18-24 hr, non-invasive μΡΕΤ/CT was performed and immediately followed by in vivo and in situ DsRed reporter-gene and NIRF optical imaging.
[0097] PET, NIRF, and DsRed Fluorescence Imaging. Fluorescence imaging was conducted using custom-built instrumentation described elsewhere (Hall et ah, 201 1). For DsRed fluorescence imaging, excitation light was supplied at 568 nm by a tunable, air- cooled, argon/krypton laser and emission light (610 ± 5 nm) was collected through a bandpass filter (Andover Corp., Salem, NH). NIRF imaging utilized excitation light at 786 nm from a laser diode and emission light was collected through two tandem bandpass filters collecting light at 830 ± 5 nm (Zhu et ah, 2010).
[0098] Non-invasive μΡΕΤ/CT was performed using an Inveon System (Siemens Medical Solutions, Knoxville, TN) with multimodality CT and docked PET. Images were analyzed and %ID/g was quantified from PET images using Inveon Research Workplace software (Siemens Medical Solutions). Target-to-background ratios (TBRs) were computed from the %ID/g normalized to normal muscle background. Previously, the inventors showed that LN DsRed fluorescence measured in situ or ex vivo provided comparable or better true- positive LN discrimination than pathology (Hall et al, 2010). Consequently, the inventors used in situ or ex vivo DsRed fluorescence as a measure of ground truth for LN cancer positivity and computed the PET true-positive rate (TPR or sensitivity) versus false-positive rate (FPR, or [1 - specificity]) of all LNs examined using ascending TBR as positive cut-off criteria during ROC curve analyses. The area under the curve (AUC) was then computed for each dual-labeled mAb tested.
[0099] Data Analyses. General statistical comparisons of mAbs were made using the Student's t-test, unpaired, or a one-way ANOVA as appropriate with Microsoft® Office Excel 2007 (Microsoft Corporation, Redmond, WA) or SigmaPlot® 11 (Systat Software, Inc., San Jose, CA) software. For determining similarity of results obtained from different imaging modalities and for performing ROC curve analyses for each mAb, SAS 9.2 (SAS Institute Inc., Cary, NC) and SigmaPlot® 1 1 (Systat Software, Inc.) software were utilized.
Example 2 - Results
[00100] Anti-EpCAM MAbs Selected by Off Rate Analysis and Competitive Binding. Screening and analyses of single-cell clones from parental wells of hybridoma fusions were performed as previously described (Pinkston et al, 20\ \; Canziani et al, 2004) and yielded a mAb panel with slower off rates than that of commercial mAb 9601 (FIG. 1). During competitive binding assays, binding of some mAbs, including mAbs 17, 73, 81, 144, and 153, to EpCAM blocked binding by mAb 9601, suggesting shared binding epitopes (FIG. 2A). However, mAbs 7, 18, 33, 38, 66, 70, 83, 85, 124, 133, and 138 did not inhibit mAb 9601 binding, suggesting epitopes that do not overlap or binding sites that do not compete with that of mAb 9601 (FIG. 2B). From subsequent analysis that compared mAb 7 to the mAb panel, the inventors found that mAbs that do not have overlapping epitope binding with mAb 9601, have an overlapping footprint with mAb 7. Thus, the mAb panel could be categorized into two unique groups with specificity for two separate, non-overlapping epitopes on the EpCAM protein. MAbs from each group, mAbs 153 and 7, were chosen for further evaluation. The VH and VL sequences, including the corresponding CDR sequences, for mAb 153 and mAb 7 are shown in Table 1. Table 1. VH and VL CDR Sequences for mAbs 153 and 7.
Figure imgf000032_0001
[00101] SPR Assessment of Anti-EpCAM MAbs 7 and 153 Fab Fragments.
Fab fragments of mAb 153 demonstrated a slower Ka or "off rate than that of mAb 7, which in turn was slower than that of mAb 9601 Fab fragment, a trend consistent with SPR analysis of dissociation avidity of captured IgG antibody using the bivalent EpCAM/Fc as the analyte (Table 2). In addition, both mAb 153 and 7 Fab fragments demonstrated significantly faster "on" rates (Ka) than that of mAb 9601. The equilibrium affinity constant (¾) was found to be 15.4, 0.54, and 2.3 nM for mAbs 9601, 153, and 7 Fab fragments, respectively. These affinity constants translated to a 28- and 7-fold increase in affinity of mAbs 153 and 7 above mAb 9601, respectively.
[00102] MAb Whole-Cell Binding Activity. As seen with mAb 9601, both mAbs 7 and 153 effectively bound PC3 cells as demonstrated by specific binding during flow cytometry (FIG. 3). An MFI fold increase of 3.9, 4.8, and 2.9 was found for cells bound by mAbs 7, 153, and 9601, respectively, relative to the MFI of cells stained with PE-conjugated secondary antibody alone. This result provides evidence that, in addition to recognizing recombinantly-expressed EpCAM/Fc, these in-house generated mAbs recognize native EpCAM on the cell surface.
[00103] Species Cross-reactivity of MAbs Tested by Whole-cell Binding. The generated panel of anti-human EpCAM mAbs bound only the human cells and did not bind to the mouse 4T1 cells at detectable levels (see FIGs. 7A and C for results from mAb 153, which are representative of those from the entire mAb panel). Conversely, rat anti-mouse EpCAM mAb G8.8 demonstrated binding to the 4T1 cells, but not to human MCF7 cells (FIGs. 7B and D).
[00104] Contrast Agent Characterization. DOTA conjugation to the mAbs occurred at varying levels for each reaction and ranged from 3.0-4.6 DOTA molecules per mAb, while conjugation of IRDye 800CW to the mAbs resulted in D/P ratios ranging from 2.1-3.4 (Table 2). Although radiolabeling reactions were performed identically, radiochemical yields were impacted by the amount of DOTA conjugated and were determined to be 60%-80%. Radiochemical purities were greater than 90% for the mAb 9601 -conjugate and greater than 95% for all other agents with typical specific activities of 0.185-0.259 MBq (5-7 μϋϊ) per μg protein.
[00105] From NIR flow cytometry, the inventors found that PC3 cells stained by IRDye-labeled mAbs 7 and 153 had significantly higher MFIs (1722.0 ± 57.4 and 1359.3 ± 94.8 MFI units, respectively) relative to PC3 cells bound by conjugated mAb 9601 (1087.0 ± 74.0 MFI units), while the MFI for mAb 7 was also significantly higher than that for mAb 153 (P < 0.05, FIG. 4). IgGi isotype control mAb 69 had a significantly lower MFI (124.7 ± 7.3 MFI units) relative to mAbs 7 and 153 (P < 0.05), as did commercial IgG2b isotype control mAb (149 ± 6.0 MFI units) relative to mAb 9601. The biological activity, measured as the mean percentage of PC3 cells stained, of conjugated mAb 7 (86.4 ± 1.8%) was significantly greater than that of IRDye-labeled mAb 9601 (79.5 ± 0.8%) (P < 0.05, Table 2). While the biological activity of mAb 153 (81.8 ± 1.5%) was also higher than that of mAb 9601, the difference was not found to be significant. The percentage of cells stained by IgGi isotype control mAb 69 (3.2 ± 0.3%) was significantly low relative to mAbs 7 and 153, as was the percentage of cells stained by commercial IgG2b isotype control mAb (0.7 ± 0.1%) relative to mAb 9601 (P < 0.05). The MFI (1 1 16.5 ± 54.5 MFI units) as well as the percentage (80.2 ± 5.5%) of cells bound by unlabeled mAb 9601 followed by incubation with IRDye-labeled secondary Ab was not significantly different from that of cells stained directly by conjugated mAb 9601, demonstrating that the NIR fluorophore label did not significantly hinder mAb binding to its target on PC3 cells.
Table 2. Characterization of Imaging Conjugates.
Figure imgf000034_0001
P < 0.05 for mAb 7 versus 9601 and 153.
**P < 0.05 for IgGi isotype control mAb 69 versus mAb 7 and 153; and for commercial IgG2b isotype control (0.7 ± 0.1%) versus mAb 9601.
†NA, not applicable as mAb 69 is an isotype control targeting pili.
[00106] Multimodal Imaging. Non-invasive, DsRed fluorescence imaging of each mouse at two-week intervals allowed for monitoring of primary prostate tumors and detection of LN involvement in some animals. While primary tumors and some metastatic LNs could be easily visualized, it was necessary to hold the abdominal skin taut on some mice for LN staging (see FIG. 8). By week 10-12 post-implantation, metastases usually involved the lumbar LNs, and sometimes also medial iliac, renal, sciatic, inguinal, or popliteal LNs. [00107] From non-invasive μΡΕΤ/CT, radiotracer signal from primary prostatic tumors and LNs bound by dual-labeled mAb was detected. FIG. 5 shows coronal (FIG. 5A), sagittal (FIG. 5B), and 3-D (FIG. 5C) views from μΡΕΤ/CT of a representative mouse in which the prostate, lumbar, and renal LNs, and one sciatic LN were PET-positive. In situ DsRed gene-reporter imaging (FIGs. 5D and G) demonstrated the presence of PC3 cells in the same tissues, confirming a primary tumor and LN metastases. DsRed fluorescence in this animal's right sciatic LN was observed during in vivo imaging of the dorsal side as well as ex vivo imaging. Similarly, in situ NIRF imaging of the same mouse (FIGs. 5E and H) demonstrated that the same cancer-positive tissue could be detected from specifically bound, dual-labeled mAb. The tissues are also shown in white light images (FIGs. 5F and I). Radiotracer and NIRF signals were also detected in the liver, kidneys, and bladder, which is not uncommon for the whole-mAb imaging agent. For all LNs examined by DsRed fluorescence and NIRF imaging, the correlation between the two imaging modalities was between 0.8 and 1.0 for all mAbs tested.
[00108] Quantitative PET Image Analyses. Table 3 lists the mean TBR of %ID/g for DsRed-positive LNs in PCa-positive mice and the AUC for each dual-labeled imaging agent tested. While the percentage of DsRed-positive (i.e. cancer-positive) LNs found in each group of mice was similar (range, 10%- 14%), the mean TBRs for DsRed- positive LNs in mice administered dual-labeled mAbs 7 (5.79 ± 3.3 %ID/g) and 153 (3.77 ± 0.9 %ID/g) were significantly higher (P < 0.05) than the mean TBR for DsRed-positive LNs in mice given the dual-labeled isotype control mAb 69 (1.79 ± 0.9 %ID/g). No significant difference was found between TBRs for in-house mAbs 7 and 153; however, the mean TBR found for mAb 9601 (3.68 ± 3.2 %ID/g) was significantly lower than that for mAb 7 (P < 0.05).
[00109] FIG. 6 shows the ROC curve generated for each dual-labeled mAb tested. The AUC for dual-labeled mAbs 7 (86.8%) and 153 (78.0%) were both significantly greater than the 50% nondiscriminatory line (P < 0.05), whereas the AUC for mAb 9601 (60.7%) was not significantly different from a 50/50 random guess (Table 3). The AUC (27%) for isotype control mAb 69, which is specific for pili expression in E. faecalis (Gao et ah, 2010), was less than 50%. Given that a perfect classification in ROC analysis is defined as TPR = 1.0 and FPR = 0, mAbs 7 and 153 each demonstrated better classification or "concordance" between μΡΕΤ/CT and DsRed-reporter gene signals, and overall higher sensitivity relative to commercial mAb 9601 and control mAb 69. Using optimal TBR cutoff values that maximized the functions of the ROC curve generated for each mAb (Table 3), the inventors found that the corresponding sensitivities for in-house mAbs 153 (94%) and 7 (93%) were substantially greater than the sensitivity found for commercial mAb 9601 (67%). The specificities were found to be similar for the three mAb-conjugates. Table 3. Quantitative PET Image Analyses using DsRed-positive LNs as the True-positive Indicator of LN Metastases.
Figure imgf000036_0001
Mean TBR of % ID/g (SD) for DsRed-positive lymph nodes in prostate cancer-positive †AUC, area under curve from receiver operating characteristic (ROC) curve analysis (curves are shown in Figure 6).
%P < 0.05 for mAbs 7 and 153 versus mAb 69, and for mAb 7 versus mAb 9601. No significant difference was found between mAbs 7 and 153.
§ < 0.05 for mAbs 7 and 153 versus AUC of 50% (line of nondiscrimination).
[00110] Comparison to anti-EpCAM mAB VU-1D9. It has also been shown that these mAbs also compare favorably to Veridex's anti-EpCAM mAb VU-1D9, a high affinity mAb that functions in rare cell detection and isolation on the Veridex FDA approved platform to quantify circulating tumor cells (CTCs) in patient blood samples. Improving the anti-EpCAM antibody reagent in this application should improve the ability to detect rare cancer cell events in patient blood samples. By using limiting dilutions of mAb 7, 153, or VU-1D9 followed by a fluorescently conjugated secondary anti-mouse antibody to label human breast cancer cell line MCF-7 cells, the inventors demonstrated improved sensitivity of mAb 7 and 153 over that of Veridex VU-1D9 (FIG. 9).
[00111] Conversion to scFvs. These lead antibody candidates were also converted into scFvs using techniques previously described (Krebber et ah, 1997). Comparison of these scFvs to the parental mAbs demonstrated that: (i) the scFvs retained affinity to natively displayed EpCAM as monitored through labeling studies of the prostate cancer cell line PC3 (FIG. 10) and (ii) the affinities of the scFvs as measured by Biacore were comparable to the measurements of Fab fragments physically cleaved from the full length mAbs 7 and 153 (FIG. 10) (Hall et ah, 2012). The conversion of the lead mAbs to scFvs with comparable affinities to the parent antibody binding domain (i) demonstrates the capability of accurately measuring and retaining affinity while converting to different antibody formats and (ii) the conversion to the scFv antibody format allows for the expression of the antibody binding domain in Escherichia coli, required for affinity maturation of this antibody using display technologies.
[00112] Cells expressing the scFv library will be incubated with the EpCAM- Fc antigen (R&D Systems) and subsequently washed to remove low affinity interactions. Those E. coli cells expressing scFv that retain EpCAM-Fc represent mutants with higher affinity to EpCAM. Detection of EpCAM-Fc levels retained on the E. coli surface is achieved with a fluorescently conjugated secondary anti-Fc antibody and cells are isolated via flow cytometry (FIG. 1 1). Selected scFvs will be expressed and their affinity determined by Biacore as and compared to parental scFv affinities (FIG. 10). To demonstrate feasibility of this system, the inventors have completed initial experiments in which both anti-EpCAM scFvs 7 and 153 have been cloned into the pAPExl vector for E. coli display. Incubation with EpCAM-Fc followed by detection with fluorescently conjugated anti-Fc demonstrated specific labeling of the E. coli population, as compared to E. coli cells expressing a control scFv with no affinity to EpCAM or labeling cells with secondary control alone (FIG. 12).
[00113] Conversion of lead anti-EpCAM scFvs to IgG and confirmation of affinity improvements. To demonstrate the activity of affinity matured antibodies head to head with commercially available anti-EpCAM mAbs including Veridex anti-EpCAM VU- 1D9 among others, the scFvs must be returned to full length IgG molecules. For IgG conversion of selected scFvs, the variable regions of the scFv will be PCR amplified. DNA encoding each of the heavy and light chain variable regions will be cloned in frame with DNA encoding the mAb constant regions. Expression will be driven by the CMV promoter in 293 Freestyle cells as previously described (An et ah, 2009). Secreted mAbs will then be purified from medium using protein A/G affinity chromatography. The concentration of purified mAbs will be determined by OD280 nm and the purity by SDS-PAGE. Biacore analysis will confirm affinity improvement of IgG as described (Hall et al, 2012).
[00114] Affinity maturation of lead anti-EpCAM antibody candidates 7 and
153. Affinity maturation of the antigen binding domain of mAb 7 and 153 will be used to achieve strikingly higher sensitivity with these antibodies for both improved diagnostic imaging performance and improvements in CTC cell detection and isolation. For affinity maturation, scFv 7 and scFv 153 DNA sequences that have been identified will be utilized as template to generate mutagenic libraries as previously described (Fromant et al. 1995). Each sequence resulting from mutagenesis that bears a unique set of mutations is then cloned into the pAPExl vector for protein expression and display of the library on the E. coli inner membrane via a new lipoprotein A (NlpA) fusion as previously described (Harvey et al, 2004; Harvey et al, 2006). [00115] Evaluation of affinity matured mAbs for cancer detection. Through limiting dilution labeling of EpCAM on prostate and breast cancer cell lines PC3 and MCF7 and subsequent flow cytometry detection, the inventors can compare the Veridex antibody VU-1D9, the parent antibodies 7 and 153, and the affinity matured candidates to monitor for improved sensitivity of cell labeling and detection. This will be done essentially as described above (FIG. 9) in the parental mAb studies. Confirmation of affinity improvements and improved sensitivity of these mAbs to epithelial carcinoma cell lines will significantly improve their utility in cancer detection for both cancer/metastatic imaging and CTC identification/isolation applications.
Example 3 - Discussion
[00116] Sentinel LN dissection (SLND) has been standard-of-care in breast cancer staging and recently, has been under investigation for staging PCa (Holl et al, 2009; Janetschek et al, 2007). It is also notable that with the advent of improved systemic treatment, recent studies have shown that breast cancer patients with limited sentinel LN metastases have gained no benefit from extensive LN dissection (Giuliano et al, 2011). Earlier detection of PCa using PSA testing and PLND may continue to decrease mortality rates due to PCa, but may also inherently increase the risk for overtreatment in low-risk PCa patients (Jemal et al, 2010), especially with the advent of new pharmacologic therapies against PCa. A molecularly targeted imaging agent that offers both benefits of noninvasive imaging and intraoperative guidance for resection of cancer-positive LNs is needed for more accurate and effective PCa staging as well as staging of other epithelial cancers.
[00117] EpCAM, which is overexpressed in >98% of PCa cases (Went et al,
2004; Rao et al, 2005), was identified 30 years ago as a tumor-associated antigen that today is known to be intensely and uniformly expressed on many epithelial carcinomas while being less intensely expressed and less accessible on the basolateral surface of normal epithelia (Benko et al, 2011 ; Herlyn et al, 1979; Sears et al, 1982; Sears et al, 1984; Stoecklein et al, 2006). When administered therapeutically, anti-EpCAM mAbs with the highest binding affinities are not well tolerated in clinical studies. Instead, anti-EpCAM mAbs with attenuated affinities exhibit less systemic toxicity and have been moderately successful in clinical trials (Munz et ah, 2010). Yet, when administered as a diagnostic at sub-therapeutic dosages, the attenuated affinity of anti-EpCAM mAbs could potentially impact imaging sensitivity, thus motivating the inventor's studies to evaluate imaging performance. Furthermore, when full-length, affinity-attenuated mAbs are converted to fragments, their reduced circulation times will not only offer more desirable pharmacokinetics to reduce background imaging signal, but would most likely result in reduced targeting and imaging sensitivity. While others have employed therapeutic mAbs as radio- and fluorescently- labeled diagnostic agents in clinical and preclinical studies, the inventors sought herein to first develop mAbs with the highest affinities and assess imaging performance prior to further modification.
[00118] The inventor's SPR-based selection strategy allowed us to identify high-affinity mAbs produced from hybridoma clones for testing in a mouse model of human PCa LN metastases. Imaging analysis was accelerated by the use of a fluorescent DsRed gene reporter as "ground truth" during assessment of LN metastases. Consistent with SPR affinity of the Fab fragment measurements, dual-labeled mAbs 153 and 7 as imaging agents possessed greater sensitivity than mAb 9601 and had a significantly higher AUC than the 50% line of non-discrimination. In contrast, the AUC of mAb 9601 was not statistically greater than the line of random chance. While the TBRs of %ID/gm of cancer-positive LNs imaged using dual-labeled mAbs 153 and 7 were significantly greater than that found using mAb 69, and while the TBR using mAb 7 was significantly greater than that found using mAb 9601, there was no significant difference between the TBRs or AUCs of dual-labeled mAbs 153 and 7, even though their measured Fab fragment affinities were different by fourfold. Because mAbs 153 and 7 possess different epitope binding footprints, the comparison of their imaging performance may not be appropriate relative to the comparison between mAbs 9601 and 153, which do share the same binding epitope on the EpCAM protein. Nonetheless, these results suggest that mAb affinity can dramatically impact imaging performance. The inventors expect that the impact of affinity on imaging performance may be more significant when using Ab fragments since limited target accessibility due to reduced circulation times could restrict imaging sensitivity.
[00119] While there are limited reports of quantitative, molecular imaging in animal models of metastatic disease in the literature, there are significant shortcomings associated with studies of Ab-based targeting of human epitopes in mouse models. Because the mAbs used herein possessed low efficiency in targeting homologous mouse EpCAM in vitro, the imaging specificity may be artificially high in these studies. On the contrary, it is also important to note that these quantitative results may have under-predicted specificity due to the fact that the mAbs tested were whole mAb molecules having Fc regions. Nonspecific binding of full mAbs by Fc receptors on immunocompetent cells, such as NK cells, macrophages, dendritic cells, and neutrophils, is inherent in animal models. Reduction of nonspecific binding can be achieved through modification of the Fc region (Strohl et al , 2009) or by developing dual-labeled specific Ab fragments, such as diabodies, or single-chain variable fragments (scFv) to eliminate Fc regions (Olafsen et al, 2010). In addition to being a mechanism for nonspecific binding (Fischman et al, 1990), Fc receptor-mediated interaction with mAb is known to play an important role in reducing mAb clearance (Kosterink et al, 2007), increasing half-life in the blood circulation (Datta-Mannan et al, 2007), and therefore increasing background signal that in turn can reduce imaging contrast. Taken together, these phenomena obviate the need for mAb fragment-based agent development during contrast agent optimization for detection of metastatic LNs in PCa.
[00120] Although preclinical NIRF tomography is under development in several laboratories, quantitative optical imaging remains to be validated and was not employed in the study presented herein. Nonetheless, fluorescent DsRed gene-reporter imaging enabled rapid identification of LN metastases and provided ground truth for assessing true- and false-positive rates for evaluation of both PET and NIRF imaging from a single, dual-labeled agent. The limit in the number of cancer cells detected from either μΡΕΤ or NIRF imaging was not assessed here, but following optimization of mAb fragment-based agent, will be studied from concordance of cytometric analyses of DsRed- and NIRF -positive cells from resected LNs. The inventors nonetheless used established, quantitative μΡΕΤ/CT imaging to assess the performance of dual-labeled mAb-based agents for detecting human PCa LN metastasis in an orthotopic mouse model. These studies demonstrate that affinity can impact imaging performance as evaluated using ROC analyses. [00121] All of the methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. More specifically, it will be apparent that certain agents which are both chemically and physiologically related may be substituted for the agents described herein while the same or similar results would be achieved. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims.
REFERENCES
The following references, to the extent that they provide exemplary procedural or other details supplementary to those set forth herein, are specifically incorporated herein by reference.
U.S. Patent Application Nos. 2002/0172677; 2004/0126828; and 20050214860
U.S. Patent Nos. 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,196,265 4,275,149
4,277,437; 4,366,241 4,742,159
4,767,720; 4,816,567 5,091,513
5,164,296; 5,196,066 5,571,698
5,627,052; 5,656,434 5,858,657
5,861,155; 5,871,907 6,406,867
6,709,659; 6,709,873
Figure imgf000042_0001
6,875,434
6,881,557; 6,891,024; and 6,946,546.
Ru et al, Cancer Res., 56, 3055-3061, 1996
Liu et al. Cell Mol. Biol, 49(2):209-216, 2003
Marks, et al, J. Biol. Chem. 267: 16007-16010 1992
Stemmer, Nature, vol. 370, p. 389-391, 1994;
Gram m a/. Proc. Natl. Acad. Set, USA, 89:3576-3580, 1992
Barbas et al, Proc. Natl. Acad. Set, USA, 91 :3809-3813, 1994
Schier et al. J. Mol. Biol. 263:551-567, 1996
Shukla-Dave et al, "Detection of prostate cancer with MR spectroscopic imaging: an expanded paradigm incorporating polyamines," Radiology. Nov 2007;245(2):499- 506.
Berney et al, International Society of Urological Pathology (ISUP) Consensus Conference on Handling and Staging of Radical Prostatectomy Specimens. Working group 4: seminal vesicles and lymph nodes, Mod Pathol. Jan 201 l;24(l):39-47.
Daskivich et al. Overtreatment of men with low-risk prostate cancer and significant comorbidity. Cancer. May 15 2011;117(10):2058-2066.
Pilepich et al, Surgical staging in carcinoma of the prostate: the RTOG experience. Radiation Therapy Oncology Group. Prostate. 1984;5(5):471-476. Musch et al, Complications of pelvic lymphadenectomy in 1,380 patients undergoing radical retropubic prostatectomy between 1993 and 2006. J Urol. Mar 2008; 179(3):923-928; discussion 928-929.
Cormier et al. , Lymphedema beyond breast cancer: a systematic review and meta-analysis of cancer-related secondary lymphedema. Cancer. Jul 27 2010.
Spizzo et al. Overexpression of epithelial cell adhesion molecule (Ep-CAM) is an independent prognostic marker for reduced survival of patients with epithelial ovarian cancer. Gynecol Oncol. Nov 2006; 103(2):483-488.
Spizzo et al. High Ep-CAM expression is associated with poor prognosis in node-positive breast cancer. Breast Cancer Res Treat. Aug 2004;86(3):207-213.
Went et al. Frequent high-level expression of the immunotherapeutic target Ep-CAM in colon, stomach, prostate and lung cancers. Br J Cancer. Jan 16 2006;94(1): 128-135. Fong et al. Ep-CAM expression in pancreatic and ampullary carcinomas: frequency and prognostic relevance. J Clin Pathol. Jan 2008;61(1):31-35.
Baeuerle EpCAM (CD326) finding its role in cancer. Br J Cancer. Feb 12 2007;96(3):417-
423.
Benko et al, Impact of the EpCAM expression on biochemical recurrence-free survival in clinically localized prostate cancer. Urol Oncol. Apr 20 2011.
Mukherjee et al. Identification of EpCAM as a molecular target of prostate cancer stroma.
Am J Pathol. Dec 2009; 175(6):2277-2287.
Edge et al, AJCC Cancer Staging Manual. Seventh ed. New York, NY: Springer; 2010. Zellweger et al. Expression patterns of potential therapeutic targets in prostate cancer. Int J
Cancer. Feb 10 2005;1 13(4):619-628.
Hall et al. Imaging prostate cancer lymph node metastases with a multimodality contrast agent. The Prostate. 201 1, DOI: 10.1002/pros.21413.
Prang et al. Cellular and complement-dependent cytotoxicity of Ep-CAM-specific monoclonal antibody MT201 against breast cancer cell lines. Br J Cancer. Jan 31
2005;92(2):342-349.
Munz et al. Side-by-side analysis of five clinically tested anti-EpCAM monoclonal antibodies. Cancer Cell Int. 2010; 10:44.
Pinkston et al. The Fsr quorum-sensing system of Enterococcus faecalis modulates surface display of the collagen-binding MSCRAMM Ace through regulation of gelE. J
Bacteriol. Sep 2011 ; 193(17):4317-4325. Canziani et al. , Kinetic screening of antibodies from crude hybridoma samples using Biacore.
Anal Biochem. Feb 15 2004;325(2):301-307.
Meares et al, Conjugation of antibodies with bifunctional chelating agents: isothiocyanate and bromoacetamide reagents, methods of analysis, and subsequent addition of metal ions. Anal Biochem. Oct 1984;142(l):68-78.
Poczatek et al. Ep-Cam levels in prostatic adenocarcinoma and prostatic intraepithelial neoplasia. J Urol. Oct 1999; 162(4): 1462-1466.
Zhu et al, Reduction of excitation light leakage to improve near- infrared fluorescence imaging for tissue surface and deep tissue imaging. Med Phys. Nov
2010;37(l l):5961-5970.
Gao et al, Enterococcus faecalis rnjB is required for pilin gene expression and biofilm formation. J Bacteriol. Oct 2010; 192(20):5489-5498.
Holl et al, Validation of sentinel lymph node dissection in prostate cancer: experience in more than 2,000 patients. Eur J Nucl Med Mol Imaging. Sep 2009;36(9): 1377-1382. Janetschek et al, [Sentinel lymph node dissection for localized prostate cancer]. Actas Urol
Esp. Jun 2007;31(6):686-692.
Giuliano et al. Axillary dissection vs no axillary dissection in women with invasive breast cancer and sentinel node metastasis: a randomized clinical trial. JAMA.
201 1;305(6):569-575.
Jemal et al, Cancer statistics, 2010. CA Cancer J Clin. Sep-Oct 2010;60(5):277-300.
Went et al. Frequent EpCam protein expression in human carcinomas. Hum Pathol. Jan
2004;35(1): 122-128.
Rao et al. Expression of epithelial cell adhesion molecule in carcinoma cells present in blood and primary and metastatic tumors. Int J Oncol. Jul 2005;27(l):49-57.
Herlyn et al, Colorectal carcinoma-specific antigen: detection by means of monoclonal antibodies. Proc Natl Acad Sci US A. Mar 1979;76(3): 1438-1442.
Sears et al. Phase-I clinical trial of monoclonal antibody in treatment of gastrointestinal tumours. Lancet. Apr 3 1982; l(8275):762-765.
Sears et al. , Effects of monoclonal antibody immunotherapy on patients with gastrointestinal adenocarcinoma. J Biol Response Mod. 1984;3(2): 138-150.
Stoecklein et al. Ep-CAM expression in squamous cell carcinoma of the esophagus: a potential therapeutic target and prognostic marker. BMC Cancer. 2006;6: 165.
Strohl Optimization of Fc-mediated effector functions of monoclonal antibodies. Curr Opin
Biotechnol. Dec 2009;20(6):685-691. Olafsen et al, Antibody vectors for imaging. Semin Nucl Med. May 2010;40(3): 167-181. Fischman et al Localization of Fc and Fab fragments of nonspecific polyclonal IgG at focal sites of inflammation. J Nucl Med. Jul 1990;31(7): 1199-1205.
Kosterink et al. Biodistribution studies of epithelial cell adhesion molecule (EpCAM)- directed monoclonal antibodies in the EpCAM-transgenic mouse tumor model. J
Immunol. Jul 15 2007; 179(2): 1362-1368.
Datta-Mannan et al. , Monoclonal antibody clearance. Impact of modulating the interaction of
IgG with the neonatal Fc receptor. J Biol Chem. Jan 19 2007;282(3): 1709-1717.
Hall et al, Imaging Prostate Cancer Lymph Node Metastases with a Multimodality Contrast
Agent. Prostate 2011.
Hall et al, Comparison of Mabs Targeting Epithelial Cell Adhesion Molecule for the Detection of Prostate Cancer Lymph Node Metastases with Multimodal Contrast Agents: Quantitative Small-Animal Pet/Ct and Nirf. J Nucl Med 2012, 53(9): 1427- 1437.
Krebber et al.., Reliable Cloning of Functional Antibody Variable Domains from
Hybridomas and Spleen Cell Repertoires Employing a Reengineered Phage Display
System. J Immunol Methods 1997, 201(l):35-55.
Fromant et al, Direct Random Mutagenesis of Gene-Sized DNA Fragments Using
Polymerase Chain Reaction. Anal Biochem 1995, 224(l):347-353.
Harvey et al, Anchored Periplasmic Expression, a Versatile Technology for the Isolation of
High-Affinity Antibodies from Escherichia Coli-Expressed Libraries. Proc Natl Acad
Sci U S A 2004, 101(25):9193-9198.
Harvey et al, Engineering of Recombinant Antibody Fragments to Methamphetamine by
Anchored Periplasmic Expression. J Immunol Methods 2006, 308(l-2):43-52.
An et al: Igg2m4, an Engineered Antibody Isotype with Reduced Fc Function. MAbs 2009, l(6):572-579.
Wallwiener et al: The Prognostic Impact of Circulating Tumor Cells in Subtypes of Metastatic Breast Cancer. Breast Cancer Res Treat 2013, 137(2):503-510.
Young et al, Circulating Tumor Cells in Lung Cancer. Acta Cytol 2012, 56(6):655-660.
Sun et al, Circulating Stem Cell-Like Epcam(+) Tumor Cells Indicate Poor Prognosis of Hepatocellular Carcinoma after Curative Resection. Hepatology 2012.

Claims

WHAT IS CLAIMED IS;
1. An isolated monoclonal antibody, wherein the antibody specifically binds to a EpCAM polypeptide and wherein the antibody competes for binding of the polypeptide with the 7 or 153 monoclonal antibody.
2. The isolated antibody of claim I, wherein the antibody competes for binding of the polypeptide with the 7 monoclonal antibody.
3. The isolated antibody of claim I, wherein the antibody competes for binding of the polypeptide with the 153 monoclonal antibody.
4. The antibody of claim I, wherein the antibody comprises:
(a) a first VH CDR at least 80% identical to VH CDR1 of 153 (SEQ ID NO: 1), or 7 (SEQ ID NO: 9);
(b) a second VH CDR at least 80% identical to VH CDR2 of 153 (SEQ ID NO: 2), or 7 (SEQ ID NO: 10);
(c) a third VH CDR at least 80% identical to VH CDR3 of 153 (SEQ ID NO: 3), or 7 (SEQ ID NO: 11);
(d) a first VL CDR at least 80% identical to VL CDR1 of 153 (SEQ ID NO: 4), or 7 (SEQ ID NO: 12);
(e) a second VL CDR at least 80% identical to VL CDR2 of 153 (SEQ ID NO: 5), or 7 (SEQ ID NO: 13); and
(f) a third VL CDR at least 80% identical to VL CDR3 of 153 (SEQ ID NO: 6), or 7 (SEQ ID NO: 14).
5. The isolated antibody of claim 4, wherein the antibody comprises:
(a) a first VH CDR at least 80% identical to SEQ ID NO: 1;
(b) a second VH CDR at least 80% identical to SEQ ID NO: 2;
(c) a third VH CDR at least 80% identical to SEQ ID NO: 3;
(d) a first VL CDR at least 80% identical to SEQ ID NO: 4;
(e) a second VL CDR at least 80% identical to SEQ ID NO: 5; and
(f) a third VL CDR at least 80% identical to SEQ ID NO: 6.
6. The isolated antibody of claim 5, wherein the antibody comprises:
(a) a first VH CDR is identical to SEQ ID NO: 1; (b) a second VH CDR is identical to SEQ ID NO: 2;
(c) a third VH CDR is identical to SEQ ID NO: 3;
(d) a first VL CDR is identical to SEQ ID NO: 4;
(e) a second VL CDR is identical to SEQ ID NO: 5; and
(f) a third VL CDR is identical to SEQ ID NO: 6.
7. The isolated antibody of claim 4, wherein the antibody comprises:
(a) a first VH CDR at least 80% identical to SEQ ID NO: 9;
(b) a second VH CDR at least 80% identical to SEQ ID NO: 10;
(c) a third VH CDR at least 80% identical to SEQ ID NO: 11 ;
(d) a first VL CDR at least 80% identical to SEQ ID NO: 12;
(e) a second VL CDR at least 80% identical to SEQ ID NO: 13; and
(f) a third VL CDR at least 80% identical to SEQ ID NO: 14.
8. The isolated antibody of claim 7, wherein the antibody comprises:
(a) a first VH CDR is identical to SEQ ID NO: 9;
(b) a second VH CDR is identical to SEQ ID NO: 10;
(c) a third VH CDR is identical to SEQ ID NO: 11 ;
(d) a first VL CDR is identical to SEQ ID NO: 12;
(e) a second VL CDR is identical to SEQ ID NO: 13; and
(f) a third VL CDR is identical to SEQ ID NO: 14.
9. The antibody of claim 4, wherein the antibody comprises:
(i) a VH domain at least about 80% identical to the VH domain of 153 (SEQ ID NO: 7) and a VL domain at least about 80% identical to the VL domain of 153 (SEQ ID NO: 8); or
(ii) a VH domain at least about 80% identical to the VH domain of 7 (SEQ ID NO: 15) and a VL domain at least about 80% identical to the VL domain of 7 (SEQ ID NO: 16).
10. The antibody of claim 9, wherein the antibody comprises a VH domain identical to the VH domain of 153 (SEQ ID NO: 7) and a VL domain identical to the VL domain of 153 (SEQ ID NO: 8).
1 1. The antibody of claim 9, wherein the antibody comprises a VH domain identical to the VH domain of 7 (SEQ ID NO: 15) and a VL domain identical to the VL domain of 7 (SEQ ID NO: 16).
12. The antibody of claim 1, wherein the antibody has an equilibrium ¾ relative to EpCAM of between about 0.1 and 10 nM.
13. The antibody of claim 12, wherein the antibody has an equilibrium ¾ relative to EpCAM of less than about 5 nM.
14. The antibody of any one of claims 1-11, wherein the antibody is the 153 or 7 antibody.
15. The antibody of any one of claims 1-1 1, wherein the antibody is recombinant.
16. The antibody of any one of claims 1-8, wherein the antibody is an IgG, IgM, IgA or an antigen binding fragment thereof.
17. The antibody of any one of claims 1-8, wherein the antibody is a Fab', a F(ab')2, a F(ab')3, a monovalent scFv, a bivalent scFv, or a single domain antibody.
18. The antibody of any one of claims 1-1 1, wherein the antibody is a human, humanized antibody or de-immunized antibody.
19. The antibody of any one of claims 1-1 1, wherein the antibody is conjugated to an imaging agent, a chemotherapeutic agent, a toxin or a radionuclide.
20. The antibody of claim 19, wherein the antibody is conjugated to a fluorophore and a radionuclide.
21. A composition comprising an antibody of any one of claims 1-11 in a pharmaceutically acceptable carrier.
22. An isolated polynucleotide molecule comprising a nucleic acid sequence encoding an antibody of any one of claims 1-1 1.
23. A recombinant polypeptide comprising an antibody VH domain comprising CDRs 1-3 of the VH domain of 153 (SEQ ID NOs: 1, 2 and 3); or CDRs 1-3 of the VH domain of 7 (SEQ ID NOs: 9, 10 and 1 1).
24. A recombinant polypeptide comprising an antibody VL domain comprising CDRs 1-3 of the VL domain of 153 (SEQ ID NOs: 4, 5 and 6); or 7 (SEQ ID NOs: 12, 13 and 14).
25. An isolated polynucleotide molecule comprising a nucleic acid sequence encoding a polypeptide of claim 23 or 24.
26. A host cell comprising one or more polynucleotide molecule(s) encoding an antibody of any one of claims 1-11 or a recombinant polypeptide of claim 23 or 24.
27. The host cell of claim 26, wherein the host cell is a mammalian cell, a yeast cell, a bacterial cell, a ciliate cell or an insect cell.
28. A method of manufacturing an antibody comprising:
(a) expressing one or more polynucleotide molecule(s) encoding a VL and VH chain of an antibody of any one of claims 1-1 1 in a cell; and
(b) purifying the antibody from the cell.
29. A method of preparing a subject for imaging, comprising administering to the subject an effective amount of an antibody of any one of claims 1-13, wherein the antibody is conjugated to an imaging agent.
30. A method of imaging a subject comprising:
(a) administering an effective amount of an antibody of any one of claims 1-13, wherein the antibody is conjugated to an imaging agent; and
(b) detecting the imaging agent in the subject.
31. The method of claim 30, wherein detecting the imaging agent comprises performing fluorescence imaging, PET, MRI, CT, SPECT or a combination thereof on the subject.
32. The method of claim 30, wherein the antibody is administered systemically.
33. The method of claim 30, wherein the imaging agent is a fluorophore, a dye, an MRI contrast agent or a radionuclide.
34. The method of claim 30, wherein the antibody is conjugated to a fluorophore and a radionuclide.
35. The method of claim 30, wherein the subject has a cancer, the method further defined as a method of detecting a cancer metastasis.
36. The method of claim 30, wherein the cancer metastasis is a lymphoid metastasis.
37. A method for detecting a cancer in a subject comprising testing for the presence of elevated level of EpCAM relative to a control in a sample from the subject, wherein the testing comprises contacting the sample with an antibody of any one of claims 1-13.
38. The method of claim 37, further defined as an in vitro method.
39. The method of claim 37, further defined as an in vivo method.
40. A method for treating a subject having a cancer comprising administering an effective amount of an antibody of any one of claims 1-13 to the subject.
41. The method of claim 40, wherein the cancer is a breast cancer, lung cancer, head & neck cancer, prostate cancer, esophageal cancer, tracheal cancer, brain cancer, liver cancer, bladder cancer, stomach cancer, pancreatic cancer, ovarian cancer, uterine cancer, cervical cancer, testicular cancer, colon cancer, rectal cancer, skin cancer or other epithelial cancer.
42. The method of claim 41, wherein the cancer is a prostate cancer.
43. The method of claim 40, wherein the antibody is administered systemically.
44. The method of claim 40, wherein the antibody is administered intravenously, intradermally, intratumorally, intramuscularly, intraperitoneally, subcutaneously, or locally.
45. The method of claim 40, wherein the antibody is conjugated to a therapeutic agent.
PCT/US2014/024021 2013-03-14 2014-03-12 Monoclonal antibodies targeting epcam for detection of prostate cancer lymph node metastases WO2014159531A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/774,795 US9790274B2 (en) 2013-03-14 2014-03-12 Monoclonal antibodies targeting EpCAM for detection of prostate cancer lymph node metastases

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361782601P 2013-03-14 2013-03-14
US61/782,601 2013-03-14

Publications (1)

Publication Number Publication Date
WO2014159531A1 true WO2014159531A1 (en) 2014-10-02

Family

ID=51625192

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/024021 WO2014159531A1 (en) 2013-03-14 2014-03-12 Monoclonal antibodies targeting epcam for detection of prostate cancer lymph node metastases

Country Status (2)

Country Link
US (1) US9790274B2 (en)
WO (1) WO2014159531A1 (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020018964A1 (en) 2018-07-20 2020-01-23 Fred Hutchinson Cancer Research Center Compositions and methods for controlled expression of antigen-specific receptors
WO2020047462A2 (en) 2018-08-30 2020-03-05 HCW Biologics, Inc. Methods of treating aging-related disorders
WO2020047333A1 (en) 2018-08-30 2020-03-05 HCW Biologics, Inc. Single-chain chimeric polypeptides and uses thereof
WO2020047299A1 (en) 2018-08-30 2020-03-05 HCW Biologics, Inc. Multi-chain chimeric polypeptides and uses thereof
WO2021163298A1 (en) 2020-02-11 2021-08-19 HCW Biologics, Inc. Methods of activating regulatory t cells
WO2021163299A1 (en) 2020-02-11 2021-08-19 HCW Biologics, Inc. Chromatography resin and uses thereof
WO2021163369A2 (en) 2020-02-11 2021-08-19 HCW Biologics, Inc. Methods of treating age-related and inflammatory diseases
WO2021222587A1 (en) 2020-04-29 2021-11-04 HCW Biologics, Inc. Anti-cd26 proteins and uses thereof
WO2021247003A1 (en) 2020-06-01 2021-12-09 HCW Biologics, Inc. Methods of treating aging-related disorders
WO2021247604A1 (en) 2020-06-01 2021-12-09 HCW Biologics, Inc. Methods of treating aging-related disorders
US20220089737A1 (en) * 2020-09-11 2022-03-24 Janssen Biotech, Inc. Multi-specific immune targeting molecules and uses thereof
US11738052B2 (en) 2019-06-21 2023-08-29 HCW Biologics, Inc. Multi-chain chimeric polypeptides and uses thereof
WO2023168363A1 (en) 2022-03-02 2023-09-07 HCW Biologics, Inc. Method of treating pancreatic cancer

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW202029980A (en) * 2018-10-26 2020-08-16 美商免疫遺傳股份有限公司 Epcam antibodies, activatable antibodies, and immunoconjugates, and uses thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070081993A1 (en) * 2003-05-31 2007-04-12 Peter Kufer Pharmaceutical composition comprising a bispecific antibody for epcam
US20090169547A1 (en) * 2005-11-24 2009-07-02 Ugur Sahin Monoclonal antibodies against claudin-18 for treatment of cancer

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070081993A1 (en) * 2003-05-31 2007-04-12 Peter Kufer Pharmaceutical composition comprising a bispecific antibody for epcam
US20090169547A1 (en) * 2005-11-24 2009-07-02 Ugur Sahin Monoclonal antibodies against claudin-18 for treatment of cancer

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
HALL ET AL.: "Comparison of mAbs Targeting Epithelial Cell Adhesion Molecule for the Detection of Prostate Cancer Lymph Node Metastases with Multimodal Contrast Agents: Quantitative Small-Animal PET/CT and NIRF", J NUCL MED., vol. 53, 7 August 2012 (2012-08-07), pages 1427 - 1437 *
RUF ET AL.: "Characterisation of the new EpCAM-specific antibody HO-3: implications for trifunctional antibody immunotherapy of cancer.", BRITISH JOURNAL OF CANCER, vol. 97, 10 July 2007 (2007-07-10), pages 315 - 321 *

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020018964A1 (en) 2018-07-20 2020-01-23 Fred Hutchinson Cancer Research Center Compositions and methods for controlled expression of antigen-specific receptors
US11401324B2 (en) 2018-08-30 2022-08-02 HCW Biologics, Inc. Single-chain chimeric polypeptides and uses thereof
US11672826B2 (en) 2018-08-30 2023-06-13 HCW Biologics, Inc. Methods of treating aging-related disorders
US11730762B2 (en) 2018-08-30 2023-08-22 HCW Biologics, Inc. Methods for stimulating proliferation or differentiation of an immune cell with a multi-chain chimeric polypeptide
WO2020047333A1 (en) 2018-08-30 2020-03-05 HCW Biologics, Inc. Single-chain chimeric polypeptides and uses thereof
US11518792B2 (en) 2018-08-30 2022-12-06 HCW Biologics, Inc. Multi-chain chimeric polypeptides and uses thereof
WO2020047462A2 (en) 2018-08-30 2020-03-05 HCW Biologics, Inc. Methods of treating aging-related disorders
WO2020047473A1 (en) 2018-08-30 2020-03-05 HCW Biologics, Inc. Single-chain and multi-chain chimeric polypeptides and uses thereof
WO2020047299A1 (en) 2018-08-30 2020-03-05 HCW Biologics, Inc. Multi-chain chimeric polypeptides and uses thereof
US11884712B2 (en) 2018-08-30 2024-01-30 HCW Biologics, Inc. Multi-chain chimeric polypeptides and uses thereof
US11738052B2 (en) 2019-06-21 2023-08-29 HCW Biologics, Inc. Multi-chain chimeric polypeptides and uses thereof
WO2021163369A2 (en) 2020-02-11 2021-08-19 HCW Biologics, Inc. Methods of treating age-related and inflammatory diseases
WO2021163299A1 (en) 2020-02-11 2021-08-19 HCW Biologics, Inc. Chromatography resin and uses thereof
WO2021163298A1 (en) 2020-02-11 2021-08-19 HCW Biologics, Inc. Methods of activating regulatory t cells
WO2021222587A1 (en) 2020-04-29 2021-11-04 HCW Biologics, Inc. Anti-cd26 proteins and uses thereof
WO2021247604A1 (en) 2020-06-01 2021-12-09 HCW Biologics, Inc. Methods of treating aging-related disorders
WO2021247003A1 (en) 2020-06-01 2021-12-09 HCW Biologics, Inc. Methods of treating aging-related disorders
US20220089737A1 (en) * 2020-09-11 2022-03-24 Janssen Biotech, Inc. Multi-specific immune targeting molecules and uses thereof
WO2023168363A1 (en) 2022-03-02 2023-09-07 HCW Biologics, Inc. Method of treating pancreatic cancer

Also Published As

Publication number Publication date
US20160024207A1 (en) 2016-01-28
US9790274B2 (en) 2017-10-17

Similar Documents

Publication Publication Date Title
US9790274B2 (en) Monoclonal antibodies targeting EpCAM for detection of prostate cancer lymph node metastases
US11548943B2 (en) Matrix metalloprotease-cleavable and serine protease-cleavable substrates and methods of use thereof
US10081680B2 (en) Anti-SIRP-alpha antibodies and bispecific macrophage enhancing antibodies
ES2778823T3 (en) Anti-CEACAM5 antibodies and their uses
ES2469369T3 (en) Humanized anti-CXCR4 antibodies for cancer treatment
ES2664989T3 (en) ANTI-CEACAM6 antibodies and their uses
US9676846B2 (en) Human monoclonal antibodies that bind insulin-like growth factor (IGF) I and II
US9737623B2 (en) Antibodies that bind activatable antibodies and methods of use thereof
EP2399934B1 (en) Novel monoclonal antibody, and use thereof
ES2615387T3 (en) Antibodies against kidney-associated antigen 1 and antigen-binding fragments of these
CN106132990A (en) Anti-MCAM antibody and associated method of use
CN115803340A (en) anti-CD 3 antibodies and uses thereof
CN114401744A (en) anti-CD 33 antibodies for the treatment of cancer
WO2022237647A1 (en) Binding molecule against dll3 and use thereof
CN111194222A (en) Compositions and methods related to xCT antibodies
ES2797089T3 (en) IGF-1R antibody and its use for cancer diagnosis
RU2816856C2 (en) Antibodies targeting epn1
US20220002435A1 (en) Antibodies targeting epn1
WO2022216672A1 (en) Anti-annexin a1 antibodies
Frigerio PSMA-specific Antibody Fragments for Prostate Cancer Imaging and Therapy
NZ736142B2 (en) Anti-pdl1 antibodies, activatable anti-pdl1 antibodies, and methods of use thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14776390

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 14774795

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14776390

Country of ref document: EP

Kind code of ref document: A1